USP17L3 antibodies are polyclonal or monoclonal reagents that bind specifically to the USP17L3 protein, a member of the ubiquitin-specific protease family. These antibodies are primarily used in research to study USP17L3's role in:
Cellular proliferation
Apoptosis regulation
Cell migration
Viral infection response
Key attributes of USP17L3 antibodies include:
| Parameter | Details |
|---|---|
| Host Species | Rabbit (most common) |
| Reactivities | Human |
| Applications | ELISA, Western Blot (WB), Immunohistochemistry (IHC) |
| Immunogen | Recombinant Human USP17L3 protein (e.g., residues 19–251 or 382–431) |
| Conjugation | Available in HRP, FITC, Biotin, or unconjugated formats |
USP17L3 is a deubiquitinating enzyme (DUB) that removes ubiquitin moieties from target proteins, modulating their stability and activity. Key functional insights:
Cancer Regulation: USP17L3 is downregulated in malignant breast cancer cells (e.g., MDA-MB-231) compared to normal or less aggressive cell lines (e.g., MCF-10A, MCF-7). Overexpression inhibits tumor growth, while knockdown enhances tumorigenesis .
ERK Signaling: USP17L3 interacts with asparaginyl endopeptidase (AEP), deubiquitinating it to suppress ERK pathway activation, thereby reducing cancer cell proliferation .
Cell Motility: While USP17 (a related isoform) regulates chemokine-induced migration via Rho GTPases , USP17L3’s direct role in motility remains under investigation.
ELISA/WB: Used to quantify USP17L3 expression in cell lysates or tissues .
Immunohistochemistry: Validated in normal and pathological tissues (e.g., breast cancer biopsies) .
Knockdown/Overexpression Models: Antibodies verify USP17L3 modulation in cell lines (e.g., MCF-7, MDA-MB-231) .
USP17L3 antibodies undergo rigorous validation:
Western Blot: Detects bands at predicted molecular weights (e.g., ~50 kDa) .
Orthogonal Validation: Confirmed via protein arrays and cross-reactivity assessments .
Specificity: CDRH3 sequence identity thresholds (>70%) ensure antigen specificity, minimizing cross-reactivity .
Isoform Specificity: USP17L3 shares homology with other USP17 family members (e.g., USP17B, USP17F), necessitating careful validation to avoid cross-reactivity .
Functional Overlap: USP17L3’s role in viral response remains underexplored compared to USP17 .
USP17L3 (ubiquitin specific peptidase 17 like family member 3) is a deubiquitinating enzyme that removes conjugated ubiquitin from specific proteins to regulate various cellular processes . It belongs to the Peptidase C19 protein family and functions by cleaving ubiquitin conjugates from targeted proteins, thereby preventing their proteasomal degradation . The canonical human USP17L3 protein has 530 amino acid residues with a molecular mass of approximately 59.5 kDa . It primarily regulates cell proliferation, cell cycle progression, apoptosis, cell migration, and cellular responses to viral infection by modifying the ubiquitination status of its target proteins . USP17L3 is predominantly localized in the nucleus and endoplasmic reticulum, where it executes its deubiquitinating functions .
Research applications for USP17L3 antibodies include several validated detection methods:
| Detection Method | Dilution Range | Compatible Antibody Formats | Common Applications |
|---|---|---|---|
| Western Blot (WB) | 1:500-1:2000 | Unconjugated, HRP-conjugated | Protein expression analysis, molecular weight verification |
| ELISA | Variable by kit | Unconjugated, HRP-conjugated, Biotin-conjugated | Quantitative protein detection, screening |
| Flow Cytometry (FCM) | Application-dependent | FITC-conjugated | Cell population analysis |
| Fluorescence Applications (FA) | Application-dependent | FITC-conjugated | Subcellular localization, co-localization studies |
Most commercially available USP17L3 antibodies have been validated using MCF-7, K562, and HepG2 whole cell lysates . When planning experiments, researchers should consider both the detection method and appropriate positive control samples to ensure reliable results.
USP17L3 antibodies require specific storage conditions to preserve their functionality and specificity. For short-term storage (up to 2 weeks), antibodies should be refrigerated at 2-8°C . For long-term storage, maintain antibodies at -20°C in small aliquots to prevent repeated freeze-thaw cycles that can degrade antibody performance . Most commercial USP17L3 antibodies are supplied in a buffer containing 50% glycerol with 0.03% Proclin 300 as a preservative . When diluting the antibody for experimental use, researchers should prepare only the volume needed for immediate use to minimize activity loss. Properly stored antibodies typically maintain activity for 12 months from the date of receipt when handled according to supplier recommendations .
USP17 (closely related to USP17L3) functions as a positive regulator of RORγt, the master transcription factor in Th17 cells, through a specific deubiquitination mechanism . The enzyme stabilizes RORγt protein expression by reducing its polyubiquitination specifically at the Lys-360 residue, thereby preventing proteasome-mediated degradation . This protective effect increases RORγt protein levels and subsequently enhances the transcriptional activation of Th17-related genes including IL-17 and IL-17F .
The mechanistic pathway operates as follows:
USP17 directly binds to RORγt in the nucleus
USP17 catalyzes the removal of ubiquitin chains from the Lys-360 residue of RORγt
Deubiquitinated RORγt escapes proteasomal degradation
Stabilized RORγt increases transcriptional activation of downstream target genes
Enhanced expression of IL-17 and other pro-inflammatory cytokines occurs
This mechanism has significant implications for autoimmune disease research, particularly for systemic lupus erythematosus (SLE), where USP17 expression is upregulated in CD4+ T cells from patients compared to healthy controls . Researchers investigating autoimmune conditions should consider USP17L3/USP17 as a potential therapeutic target to modulate RORγt-mediated pathways in Th17 cells, potentially reducing pathogenic inflammation in conditions like SLE .
USP17L3/USP17 plays a critical role in regulating Ras activation and subsequent cell proliferation through its interaction with the Ras processing pathway . Research has demonstrated that USP17 disrupts Ras plasma membrane localization and activation, leading to marked inhibition of cell proliferation . The mechanism involves USP17 blocking RCE1 (Ras converting enzyme 1) activity, which is essential for proper Ras processing and membrane localization .
The disruption of Ras signaling occurs through the following cascade:
USP17 expression interferes with RCE1 enzymatic function
Impaired RCE1 activity prevents proper post-translational modification of Ras proteins
Incompletely processed Ras fails to localize correctly to the plasma membrane
Mislocalized Ras cannot interact effectively with upstream activators and downstream effectors
The compromised Ras activation leads to inhibition of proliferative signaling pathways
This relationship has significant implications for cancer research models where Ras signaling is frequently dysregulated. When designing experiments to investigate USP17L3/USP17 in cancer contexts, researchers should incorporate analyses of Ras localization, activation status (GTP-bound Ras), and downstream signaling effects in addition to proliferation assessments . This multi-parameter approach provides more comprehensive insights into how USP17L3/USP17 modulates oncogenic signaling networks.
Detecting endogenous USP17L3 in primary human cells presents several technical challenges due to its regulation, expression patterns, and structural characteristics:
To overcome these challenges, researchers should implement a multi-faceted approach. For immunoblotting applications, use cytokine stimulation protocols (such as IL-4 or IL-6) to enhance USP17L3 expression before cell lysis . For immunofluorescence detection, combine subcellular fractionation with confocal microscopy to accurately localize USP17L3. Additionally, complementary techniques such as RT-qPCR using the validated primer pairs (5′-GAGCACTTGGTGGAAAGAGC-3′ and 5′-TGATGGTTCTTCATCCCACA-3′) can verify expression at the transcript level before proceeding to protein detection .
Proper control design is critical for USP17L3 antibody validation experiments to ensure specificity and reliability of results:
For overexpression controls, researchers can use the validated RORγt and USP17 constructs previously documented in the literature, which can be amplified by PCR with human cDNA from HEK293T cells and cloned into tagged vectors (FLAG, Myc, or HA-tagged) . When planning knockdown experiments, researchers should design siRNA or shRNA targeting USP17L3 specifically, with validation using RT-qPCR with the primers: 5′-GAGCACTTGGTGGAAAGAGC-3′ (forward) and 5′-TGATGGTTCTTCATCCCACA-3′ (reverse) .
USP17L3 localizes primarily to the nucleus and endoplasmic reticulum, requiring specialized sample preparation approaches for optimal detection:
For Western blot applications, researchers should prepare fresh lysates due to potential degradation of USP17L3 during storage. When performing immunofluorescence microscopy, a brief pre-permeabilization step with 0.01% digitonin before fixation can improve antibody access to nuclear and ER compartments without disrupting subcellular structures. For co-localization studies, include appropriate markers such as Lamin B1 (nuclear envelope) or Calnexin (ER) alongside USP17L3 detection to confirm proper compartmentalization.
Measuring USP17L3 deubiquitinating enzyme (DUB) activity requires specialized approaches beyond simple protein detection:
| Activity Assay | Methodology | Applications |
|---|---|---|
| Fluorogenic Substrate Assay | Measure cleavage of ubiquitin-AMC or Ub-Rhodamine | Quantitative analysis of catalytic activity |
| Di-ubiquitin Cleavage Assay | Analyze cleavage of specific di-ubiquitin linkages (K48, K63, etc.) | Determine linkage preference |
| Cellular Substrate Deubiquitination | Co-IP of substrate (e.g., RORγt) followed by ubiquitin immunoblotting | Assess activity toward physiological substrates |
| Reporter-based Assay | Ub-X-GFP fusion constructs with fluorescence readout | Cell-based screening applications |
| Enzymatic Inhibition Assay | Activity measurement in presence of DUB inhibitors | Confirm specificity of observed activity |
For the most physiologically relevant assessment, researchers should utilize the cellular substrate deubiquitination approach. This involves immunoprecipitating potential substrates like RORγt and assessing their ubiquitination status in the presence or absence of USP17L3 expression or activity . When using the catalytically inactive mutant USP17C89S as a negative control, researchers can distinguish between enzymatic and non-enzymatic effects on substrate proteins .
To establish a functional reporter system, the validated luciferase reporter assay using the Il17a promoter can be employed. This involves cotransfecting the Il17a luciferase reporter plasmid with USP17L3 expression constructs and measuring the resulting luciferase activity, which correlates with USP17L3's functional impact on transcriptional regulation .
USP17L3 antibodies provide valuable tools for investigating its role in autoimmune disease models, particularly given the established connection between USP17 and systemic lupus erythematosus (SLE) . Researchers can implement several approaches:
| Investigation Approach | Methodology | Data Interpretation Guidelines |
|---|---|---|
| Expression Analysis in Patient Samples | Immunoblotting or immunohistochemistry of tissues/PBMCs from autoimmune patients vs. controls | Compare band intensity normalized to loading controls; quantify across patient cohorts |
| Th17 Cell Population Studies | Flow cytometry with USP17L3 and RORγt co-staining | Analyze correlation between USP17L3 levels and Th17 markers in disease states |
| Functional Impact Assessment | siRNA knockdown of USP17L3 in patient-derived T cells followed by cytokine profiling | Measure changes in IL-17, IL-17F and other Th17-related cytokines after USP17L3 depletion |
| Intervention Studies | Treatment of cells with DUB inhibitors targeting USP17L3 | Monitor effects on RORγt stability and downstream inflammatory markers |
When interpreting data from these studies, researchers should consider that USP17L3 expression is up-regulated in CD4+ T cells from SLE patients compared to healthy controls . This differential expression suggests that USP17L3 could serve as both a biomarker and a therapeutic target. The correlation between USP17L3 levels, RORγt stability, and IL-17 production provides a mechanistic framework for understanding how USP17L3 contributes to autoimmune pathogenesis.
For validating experimental findings, researchers should use the established qPCR primers: IL-17A (5′-ACCAATCCCAAAAGGTCCTC-3′ and 5′-GGGGACAGAGTTCATGTGGT-3′), IL-17F (5′-CCTCCCCCTGGAATTACACT-3′ and 5′-ACCAGCACCTTCTCCAACTG-3′), RORγt (5′-CTGCTGAGAAGGACAGGGAG-3′ and 5′-AGTTCTGCTGACGGGTGC-3′), and USP17 (5′-GAGCACTTGGTGGAAAGAGC-3′ and 5′-TGATGGTTCTTCATCCCACA-3′) .
Recent research has implicated USP17L3 in lipid metabolism and lipid droplet (LD) biology, providing new avenues for investigation:
| Research Approach | Methodology | Expected Outcomes |
|---|---|---|
| LD Proteomics Analysis | Mass spectrometry of isolated lipid droplet fractions | Identification of USP17L3 association with LDs and potential interaction partners |
| Functional LD Assays | Analysis of LD parameters after USP17L3 knockdown/overexpression | Quantitative changes in LD number, size, dispersion, shape, and intensity |
| Rescue Experiments | Reintroduction of wild-type vs. catalytically inactive USP17L3 | Determination of whether enzymatic activity is required for LD effects |
| Substrate Identification | IP-MS or BioID proximal labeling coupled with USP17L3 | Discovery of LD-associated proteins regulated by USP17L3 deubiquitination |
When designing these experiments, researchers should consider the established parameters for lipid droplet analysis: number (LDs per cell), dispersion (percent touching), shape (eccentricity), size (median radius), and intensity (integrated intensity) . These metrics provide a comprehensive profile of how USP17L3 impacts lipid droplet biology.
To facilitate data interpretation, researchers should employ high-content microscopy analysis with appropriate staining protocols (such as BODIPY for neutral lipids) and conduct parallel biochemical assays to measure cellular triglyceride content. The integration of imaging and biochemical data provides a more comprehensive understanding of USP17L3's role in lipid metabolism regulation.
Researchers may encounter contradictory results when studying USP17L3, particularly regarding its effects on cell proliferation and its substrate specificity. These apparent contradictions can be systematically addressed:
| Contradiction Type | Validation Approach | Resolution Strategy |
|---|---|---|
| Cell Type-Specific Effects | Parallel testing in multiple cell types | Map cell type dependencies and identify contextual factors |
| Substrate Specificity Discrepancies | Direct comparison of deubiquitination efficiency toward different substrates | Determine hierarchy of substrates and regulatory mechanisms |
| Opposing Functional Outcomes | Time-course experiments with inducible expression systems | Identify biphasic responses or temporal regulation patterns |
| Technical Artifacts | Antibody validation using multiple detection methods | Ensure reagent specificity and appropriate experimental conditions |
When addressing contradictory findings regarding USP17L3's impact on proliferation, researchers should note that while USP17 has been shown to inhibit cell proliferation by disrupting Ras plasma membrane localization , its effect on RORγt stability could promote proliferation of specific T cell subsets . This apparent contradiction may reflect cell type-specific functions or pathway-dependent effects.
To validate experimental findings, researchers should implement multiple complementary approaches. For example, when studying USP17L3's impact on a potential substrate, combine in vitro deubiquitination assays, co-immunoprecipitation studies, and functional readouts in cellular models. The use of catalytically inactive mutants (USP17C89S) as negative controls is essential for confirming the enzymatic specificity of observed effects .
Several emerging technologies hold promise for overcoming current limitations in USP17L3 research:
| Technology | Application to USP17L3 Research | Potential Impact |
|---|---|---|
| CRISPR-Cas9 Gene Editing | Generation of endogenous tagged USP17L3 cell lines | Study of physiological expression levels and dynamics |
| Proximity Labeling (BioID/TurboID) | Identification of USP17L3 interactome in living cells | Discovery of novel substrates and regulatory partners |
| Single-Cell Proteomics | Analysis of USP17L3 expression heterogeneity | Understanding of cellular subpopulations and regulation |
| Cryo-EM Structural Analysis | Determination of USP17L3 structure with substrates | Design of specific inhibitors and mechanistic insights |
| Organoid Models | Study of USP17L3 function in tissue-specific contexts | Translation of findings to physiologically relevant systems |
The integration of these technologies could significantly advance our understanding of USP17L3 biology. For example, combining CRISPR-mediated endogenous tagging with live-cell imaging would enable researchers to monitor USP17L3 dynamics during cellular processes such as cell cycle progression, cytokine stimulation, or metabolic stress. Similarly, applying proximity labeling approaches would help identify the USP17L3 interactome under different cellular conditions, potentially revealing context-specific substrates and regulatory mechanisms.
Researchers should consider developing collaborative approaches that leverage these complementary technologies to build a more comprehensive understanding of USP17L3 function across different biological contexts.