The USP17L3 Antibody, FITC Conjugated is a fluorescently labeled polyclonal antibody designed for the detection of Ubiquitin carboxyl-terminal hydrolase 17-like protein 3 (USP17L3), a deubiquitinating enzyme involved in regulating cellular processes such as proliferation, apoptosis, and viral response . Conjugation with fluorescein isothiocyanate (FITC) enables visualization in fluorescence-based assays, including immunofluorescence (IF) and flow cytometry .
Target:
USP17L3 (UniProt ID: A6NCW0), a member of the ubiquitin-specific protease family, removes ubiquitin moieties from substrates to modulate protein degradation and signaling .
FITC conjugation involves covalent binding of the fluorophore to primary amines (lysine residues) on the antibody. Key parameters include:
FITC-to-Antibody Ratio: Typically 3–6 FITC molecules per IgG molecule .
Optimal Conditions: pH 9.5, 25 mg/ml antibody concentration, and 30–60 minutes at room temperature .
Purification: Antigen affinity chromatography ensures specificity, with final formulation in PBS containing 50% glycerol and 0.03% ProClin-300 .
Critical Notes:
This antibody is validated for:
Immunofluorescence (IF): Detects USP17L3 in fixed/permeabilized cells at recommended dilutions (e.g., 1:500 in PBS/10% FBS) .
Western Blot (WB): Identifies USP17L3 (~30–35 kDa) in human cell lysates .
Research Findings:
USP17L3 is a deubiquitinating enzyme that removes conjugated ubiquitin from specific proteins to regulate different cellular processes . As a member of the ubiquitin-specific protease family, it plays critical roles in protein degradation pathways, cell cycle regulation, and potentially in disease mechanisms. Researchers study USP17L3 to understand its contributions to cellular homeostasis and potential therapeutic implications in conditions where ubiquitin-mediated processes are dysregulated.
Most commercially available USP17L3 antibodies with FITC conjugation share these specifications:
For optimal staining with USP17L3-FITC antibody, follow these methodological steps:
Begin with a cell viability check - ensure viability is >90% as dead cells can lead to high background scatter and false positive staining .
Use appropriate cell numbers - a concentration of 10^5 to 10^6 cells is recommended to avoid clogging the flow cell and obtain good resolution .
If studying USP17L3, which is likely intracellular, cells will require fixation and permeabilization. Standard protocols using 2-4% paraformaldehyde for fixation followed by a gentle permeabilization agent (0.1-0.5% saponin or Triton X-100) are recommended .
Block non-specific binding sites using appropriate blockers:
Perform all staining steps on ice to prevent internalization of membrane antigens, and consider using PBS with 0.1% sodium azide .
For rigorous experimental design with USP17L3-FITC antibody, incorporate these essential controls:
Unstained cells control: Measures autofluorescence from endogenous fluorophores that may increase the false positive rate .
Negative cells control: If available, cell populations known not to express USP17L3 should be used to confirm primary antibody specificity .
Isotype control: Use a rabbit polyclonal IgG conjugated to FITC with no known specificity for your target, at the same concentration as your USP17L3 antibody. This assesses background staining due to Fc receptor binding .
Secondary antibody control: If using indirect staining methods, include cells treated only with secondary fluorophore-conjugated antibody to detect non-specific binding .
Single-stain controls: If performing multicolor flow cytometry, these are essential for calculating compensation matrices to correct for spectral overlap of fluorophores .
The appropriate controls enable accurate interpretation of flow cytometry data and help distinguish specific from non-specific binding events.
Optimization of USP17L3-FITC antibody staining requires methodical adjustment of several parameters:
Titration of antibody concentration: Perform a dilution series (e.g., 1:50, 1:100, 1:200, 1:500) to determine the optimal concentration that yields the highest signal-to-noise ratio. Calculate the staining index for each concentration:
Incubation time and temperature: Test different combinations (e.g., 30 min at 4°C, 60 min at 4°C, 15 min at room temperature) to find optimal conditions.
Fixation and permeabilization protocol adjustments: Different cell types may require modified protocols:
Blocking optimization: Test different blocking reagents (BSA, casein, commercial blocking solutions) at various concentrations to reduce background.
Cell type-specific considerations: For immune cells, more aggressive Fc receptor blocking might be necessary. For tissues with high autofluorescence (e.g., liver, brain), additional steps to reduce background might be required .
Document all optimization steps systematically to establish a reproducible protocol that works specifically for your experimental system.
Proper compensation is critical when using FITC-conjugated antibodies in multicolor panels due to spectral overlap:
Prepare single-stain controls: For each fluorochrome in your panel, prepare a sample stained with only that fluorochrome. For FITC, use your USP17L3-FITC antibody on a positive control sample .
Calculate the spillover matrix: Most modern flow cytometry software can automatically calculate the spillover matrix, but understanding the principles is important. The matrix represents the fraction of one fluorochrome's signal detected in other channels . For example, a typical spillover matrix might look like:
1.004 & -0.123 & -0.014 \\ -0.032 & 1.004 & 0.000 \\ 0.000 & 0.000 & 1.000 \end{pmatrix} $$ This matrix shows how fluorochromes (FITC, PE, APC) spill into each other's detection channels[4].Apply compensation corrections: The true fluorescent intensity (T) can be calculated from observed intensities (O) using:
Where f is the spillover matrix and the indices represent parameters and experiments .
Verify compensation: After applying the calculated compensation, check your control samples to ensure populations are properly aligned along their axes without diagonal skewing, which would indicate under or over-compensation .
Consider fluorescence-minus-one (FMO) controls: These are particularly important when identifying populations with subtle differences in expression levels .
When analyzing USP17L3 expression in mixed cell populations, consider these advanced approaches:
Data-driven gating strategies: Rather than arbitrary gates, use statistically-based data-driven thresholds. Define positive expression as the level for which 95% of unstained cells show lower expression .
Dimensionality reduction techniques: For complex datasets:
Principal Component Analysis (PCA) can help identify correlations between USP17L3 expression and other markers
Example PCA scoring coefficients for a three-parameter analysis might be:
| Variable | PC1 | PC2 | PC3 |
|---|---|---|---|
| USP17L3 | 0.711 | -0.624 | 0.324 |
| Marker 2 | -0.566 | -0.722 | -0.398 |
| Marker 3 | 0.418 | -0.298 | 0.859 |
The interpretation would be that 50.6% (100×0.711²) of variance in USP17L3 expression is represented in PC1 .
Cell subset identification: For identifying specific subpopulations expressing USP17L3:
Use hierarchical gating based on lineage markers before analyzing USP17L3 expression
Consider density-based clustering algorithms which can reveal subpopulations automatically
Calculate the ratio of marginalized density of a particular aliquot relative to the marginalized density of a negative control
Statistical rigor: Apply appropriate statistical tests when comparing USP17L3 expression between different experimental conditions, considering whether parametric or non-parametric tests are appropriate based on your data distribution .
When facing staining issues with USP17L3-FITC antibody, employ this systematic troubleshooting approach:
Weak signal problems:
Verify target expression in your cell type using alternative methods (Western blot, qPCR)
Check antibody quality by testing FITC fluorescence directly using anti-FITC antibodies
Increase antibody concentration after performing proper titration
Optimize fixation and permeabilization protocols; some epitopes are fixation-sensitive
For intracellular targets like USP17L3, ensure adequate permeabilization
Consider signal amplification methods if expression is naturally low
High background/nonspecific staining:
Implement more stringent blocking (5-10% serum, commercial blocking buffers)
Include Fc receptor blocking when working with cells like leukocytes
Reduce antibody concentration to minimize nonspecific binding
Check for spillover from other fluorochromes if using multiple markers
Ensure cells are properly washed between steps
Test with isotype control to identify sources of nonspecific binding
FITC-specific considerations:
FITC is sensitive to pH; ensure buffers are maintained at optimal pH (7.2-7.4)
FITC is susceptible to photobleaching; minimize light exposure during preparation
Consider fluorescence loss if samples cannot be analyzed immediately
If working in tissues with high autofluorescence, consider alternative fluorophores with different spectral properties
Antibody-specific issues:
Effective multiplexing with USP17L3-FITC antibody requires careful panel design and validation:
Strategic panel design:
Place USP17L3-FITC on the appropriate laser line (488nm) for optimal excitation
Pair with fluorophores that have minimal spectral overlap with FITC (e.g., APC, PE-Cy7)
Consider brightness hierarchy - if USP17L3 expression is low, FITC may not be the optimal choice as it has moderate brightness compared to newer fluorophores
Limit the total number of FITC-conjugated antibodies in your panel to one to avoid compensation challenges
Validation steps for multiplex panels:
Perform single-stain controls for each marker
Include fluorescence-minus-one (FMO) controls for each channel
Test antibody combinations to identify any unexpected interactions
Validate staining patterns match known biological distributions for each marker
Advanced multiplexing strategies:
Consider sequential staining protocols for complex panels
Implement barcoding approaches for high-dimensional analysis
When analyzing data from high-parameter experiments, employ dimensionality reduction techniques like tSNE or UMAP to visualize relationships between markers
Utilize clustering algorithms to identify novel cell populations based on multiple marker expression patterns
FITC-specific multiplexing considerations:
FITC has relatively broad emission that can spill into other channels
Use spill index calculations to determine the impact on neighboring detectors:
In panels with many markers, consider whether a brighter alternative to FITC (such as Alexa Fluor 488) might be preferable for detecting USP17L3 if expression levels are low
While primarily validated for ELISA applications, researchers may adapt USP17L3-FITC antibodies for imaging with these considerations:
Microscopy-specific optimization:
FITC has an excitation maximum around 495nm and emission maximum around 525nm, requiring appropriate filter sets
FITC is susceptible to photobleaching, so consider anti-fade mounting media containing agents like DABCO or ProLong Gold
Titrate antibody concentrations specifically for microscopy applications, which often differ from flow cytometry concentrations
Background autofluorescence may be more problematic in tissues; perform appropriate controls
Sample preparation modifications:
For tissue sections, optimize antigen retrieval methods if necessary
For adherent cells, test different fixation protocols (paraformaldehyde, methanol, acetone) to determine which best preserves USP17L3 epitopes
Consider detergent concentration carefully - excessive permeabilization can disrupt cellular architecture
For co-localization studies, sequential staining may be necessary to avoid antibody cross-reactivity
Advanced imaging considerations:
If working with thick tissue sections, confocal microscopy may be necessary to resolve USP17L3 localization
Super-resolution techniques may require additional validation as FITC's photophysical properties are not optimal for all super-resolution approaches
For live-cell imaging, consider that USP17L3-FITC antibodies would require microinjection or other specialized delivery methods
Quantitative image analysis requires consistent acquisition parameters and appropriate controls
Alternative approaches if direct antibody application fails:
Consider creating expression constructs for USP17L3-GFP fusion proteins for live-cell studies
Indirect immunofluorescence using primary non-conjugated USP17L3 antibody and secondary FITC-conjugated anti-rabbit IgG
Proximity ligation assays for studying interactions between USP17L3 and potential partner proteins
Investigating protein-protein interactions involving USP17L3 requires specialized approaches:
Co-immunoprecipitation followed by flow analysis:
Perform co-IP using anti-USP17L3 antibody
Analyze precipitates using flow cytometry with FITC-conjugated USP17L3 antibody and other markers
Compare interaction patterns under different cellular conditions
Protein proximity assays:
Use USP17L3-FITC antibody in combination with other protein-specific antibodies conjugated to compatible fluorophores
Implement Förster Resonance Energy Transfer (FRET) analysis:
FITC can serve as a donor fluorophore
Pair with an appropriate acceptor fluorophore attached to antibodies against potential interaction partners
Energy transfer occurs only when proteins are in close proximity (<10nm)
Calculate FRET efficiency using:
where F_DA is donor fluorescence in presence of acceptor and F_D is donor fluorescence alone
Pathway analysis using phospho-specific antibodies:
Design panels combining USP17L3-FITC with antibodies against phosphorylated signaling molecules
Analyze how USP17L3 expression correlates with activation of specific pathways
Compare pathway activation patterns in cells with normal versus altered USP17L3 expression
Kinetic studies of protein interactions:
Use flow cytometry to track changes in USP17L3 associations over time following stimulation
Generate temporal profiles of protein complex formation and dissociation
Correlate with functional cellular outcomes
For rigorous validation of USP17L3-FITC antibody specificity, implement these methodological approaches:
Genetic validation approaches:
Compare staining in wild-type cells versus USP17L3 knockout cells (CRISPR/Cas9 generated)
Use siRNA/shRNA knockdown to reduce USP17L3 expression and confirm corresponding reduction in antibody signal
Overexpress USP17L3 and verify increased antibody binding proportional to expression level
Biochemical validation:
Perform peptide competition assays using the immunogen peptide (amino acids 19-251 of USP17L3)
Pre-incubate antibody with increasing concentrations of blocking peptide before cell staining
Quantify dose-dependent signal reduction to confirm epitope specificity
Western blot analysis to confirm the antibody recognizes a protein of the correct molecular weight
Cross-platform validation:
Correlate flow cytometry results with data from orthogonal methods:
Western blotting
Immunohistochemistry
Mass spectrometry identification of USP17L3
Compare results from multiple antibody clones recognizing different USP17L3 epitopes
Species and isoform specificity testing:
USP17L3 functions as a deubiquitinating enzyme that removes conjugated ubiquitin from specific proteins . Research into these processes can be approached through:
Cell cycle-dependent expression analysis:
Synchronize cells at different cell cycle stages
Use USP17L3-FITC antibody in flow cytometry combined with DNA content analysis
Correlate USP17L3 expression with specific cell cycle phases
Create expression profiles showing how USP17L3 levels fluctuate throughout the cell cycle
Deubiquitination activity correlation:
Treat cells with proteasome inhibitors (MG132, bortezomib)
Analyze changes in USP17L3 expression and localization
Combine with ubiquitin antibody staining to assess correlation between USP17L3 levels and total ubiquitinated protein levels
Substrate identification approaches:
Use USP17L3-FITC to sort cells with different expression levels
Perform proteomics analysis on sorted populations to identify differentially ubiquitinated proteins
Validate candidate substrates using co-immunoprecipitation followed by ubiquitin Western blotting
Stress response studies:
Subject cells to various stressors (oxidative stress, ER stress, hypoxia)
Measure changes in USP17L3 expression using the FITC-conjugated antibody
Correlate with cellular stress response markers to understand functional relevance
Working with USP17L3-FITC antibodies across different cellular systems requires specific methodological adaptations:
Sample preparation differences:
Primary cells often require gentler isolation procedures to maintain viability
Cell lines may need authentication to confirm identity before USP17L3 analysis
Primary cells typically show higher variability between donors, requiring larger sample sizes
Consider density gradient separation for specific primary cell populations before antibody staining
Optimization parameters:
| Parameter | Primary Cells | Cell Lines |
|---|---|---|
| Cell number starting material | Higher (≥10^7) | Lower (10^6) |
| Fixation sensitivity | Often more sensitive | Generally robust |
| Autofluorescence | Typically higher | Usually lower |
| Fc receptor blocking | Critical for immune cells | Less critical for many lines |
| Donor variability | High | Low |
| Passage effects | N/A | Must be controlled |
Functional correlations:
For primary cells, correlate USP17L3 expression with donor characteristics or disease states
For cell lines, compare expression across panels representing different tissue types or genetic backgrounds
Consider how culture conditions affect USP17L3 expression in both systems
Primary cells may require fresh analysis, while cell lines can often be fixed and stored
Validation requirements:
Primary cell experiments typically require multiple donors to establish patterns
Cell line work benefits from testing across multiple related lines
Consider genetic manipulation validation approaches based on cell type:
Primary cells: transient siRNA, viral vectors
Cell lines: stable knockout/knockdown systems
Emerging technologies offer new possibilities for USP17L3 research beyond current applications:
Mass cytometry (CyTOF) integration:
While direct FITC conjugates aren't used in CyTOF, principles from flow cytometry panels can inform metal-tagged antibody panel design
Correlation between traditional flow cytometry USP17L3-FITC data and CyTOF data using metal-tagged USP17L3 antibodies
High-dimensional analysis of USP17L3 in relation to dozens of other parameters simultaneously
Spatial profiling technologies:
Multiplex immunofluorescence allows simultaneous detection of USP17L3 alongside multiple markers in tissue context
Digital spatial profiling technologies can quantify USP17L3 expression with spatial resolution
Single-cell spatial transcriptomics can correlate USP17L3 protein expression with transcriptional profiles in situ
Single-cell multi-omics approaches:
CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) could incorporate USP17L3 antibodies
Correlation of USP17L3 protein expression with transcriptome and epigenetic features at single-cell resolution
Integration of protein, RNA, and chromatin accessibility data to understand USP17L3 regulation
Advanced flow cytometry approaches:
Spectral flow cytometry allows better resolution of FITC from spectrally similar fluorophores
Imaging flow cytometry combines morphological information with expression data
High-throughput flow cytometry for screening compounds that modulate USP17L3 expression or function
Understanding USP17L3's roles in disease could guide therapeutic approaches:
Cancer research applications:
Analyze USP17L3 expression across patient-derived tumor samples
Correlate expression patterns with clinical outcomes and treatment responses
Investigate whether USP17L3 contributes to cancer progression through deubiquitination of key oncogenic proteins
Evaluate USP17L3 as a potential therapeutic target or biomarker
Immune system regulation:
Study USP17L3 expression in various immune cell subsets
Investigate changes in USP17L3 levels during immune activation
Analyze potential roles in cytokine signaling through deubiquitination of pathway components
Explore connections to autoimmune or inflammatory conditions
Developmental processes:
Track USP17L3 expression during cellular differentiation
Study potential roles in stem cell maintenance through protein stabilization
Investigate functions in embryonic development if animal models are developed
Therapeutic development considerations:
USP17L3-FITC antibodies could be used to screen for compounds that modulate its expression
Development of therapeutic antibodies would require extensive validation beyond research-grade reagents
Monitoring USP17L3 levels could potentially serve as a biomarker for treatment efficacy in certain conditions
By systematically investigating these aspects of USP17L3 biology, researchers can expand our understanding of this deubiquitinating enzyme and potentially identify novel therapeutic approaches for diseases involving ubiquitin-mediated processes.