OTUB1 regulates immune responses through two mechanisms:
Canonical deubiquitinase activity: Stabilizes proteins by removing K48- or K63-linked polyubiquitin chains .
Non-canonical activity: Inhibits E2 ubiquitin-conjugating enzymes to block ubiquitination .
CD8+ T cell and NK cell activation: OTUB1 suppresses IL-15-mediated AKT phosphorylation, acting as a checkpoint for T cell and NK cell priming .
NF-κB signaling in dendritic cells (DCs): Stabilizes UBC13, enhancing TRAF6 and IRAK1 ubiquitination to amplify cytokine production (e.g., IL-6, TNF) .
Influenza A Virus (IAV): OTUB1 stabilizes the viral NS2 protein, promoting viral RNA synthesis and replication. NS2 ubiquitination destabilizes the protein, but OTUB1 counteracts this to enhance virion production .
Immune evasion: IAV NS1 induces OTUB1 degradation to suppress RIG-I-dependent antiviral signaling .
OTUB1 is overexpressed in multiple cancers (e.g., breast, colorectal) and promotes tumor progression by:
The OTUB1 antibody has been experimentally validated in diverse models:
Targeting OTUB1 offers potential strategies for:
OTUB1 (OTU deubiquitinase, ubiquitin aldehyde binding 1) is a highly specific deubiquitinating enzyme that preferentially cleaves 'Lys-48'-linked conjugated ubiquitin from proteins, playing a crucial regulatory role in protein turnover by preventing degradation . As a member of the Peptidase C65 protein family, OTUB1 functions as a hydrolase and is widely expressed across multiple tissue types . Its significance extends to immune responses, DNA damage repair pathways, and various disease mechanisms, making it a valuable target for research across multiple disciplines . Understanding OTUB1's dual mechanisms - both canonical (catalytic) and non-canonical - provides insight into cellular regulatory networks that control protein stability and signaling pathways.
When selecting an OTUB1 antibody, researchers should consider several critical factors to ensure experimental success:
Isoform specificity: OTUB1 exists in two isoforms - a ubiquitously expressed 31 kDa form and a more restricted 35 kDa isoform (ARF-1) primarily found in lymphoid organs . Verify whether the antibody detects one or both isoforms.
Species reactivity: Confirm the antibody's validated reactivity with your species of interest. Many OTUB1 antibodies show cross-reactivity with human, mouse, and rat samples , but this should be verified for your specific application.
Application validation: Select antibodies specifically validated for your intended application. For instance, some antibodies perform well in Western blot but may not be validated for immunohistochemistry or immunoprecipitation .
Domain recognition: Consider which domain or epitope of OTUB1 the antibody targets, particularly if you are investigating specific protein interactions or modifications .
Validation methods: Look for antibodies with enhanced validation using techniques such as RNAi knockdown, which provides stronger evidence of specificity .
OTUB1's two isoforms have distinct expression patterns and potentially different functions, which necessitates careful antibody selection . The shorter 31 kDa isoform is ubiquitously expressed while the longer 35 kDa isoform (ARF-1) shows more restricted expression primarily in lymphoid organs . This distinction has significant experimental implications:
Functional differences: The isoforms may have opposing regulatory effects. For example, in T-cell anergy, isoform 1 destabilizes RNF128/GRAIL (preventing anergy), while isoform 2 stabilizes RNF128 (promoting anergy) .
Antibody epitope consideration: Verify whether your antibody can distinguish between isoforms or detects both. This is especially important when studying tissues or cell types where both isoforms might be present .
Western blot interpretation: When performing western blot analysis, be prepared to observe bands at both 31 kDa and 35 kDa, and understand which isoform(s) you are detecting .
Experimental controls: Consider including controls that express only one isoform to validate antibody specificity and to correctly interpret results from tissues expressing multiple isoforms .
For optimal Western blot detection of OTUB1, consider the following methodological recommendations:
For effective immunofluorescence detection of OTUB1:
Fixation method: Use 4% paraformaldehyde for 10-15 minutes at room temperature, which preserves OTUB1 epitope accessibility while maintaining cellular architecture.
Permeabilization: Since OTUB1 is primarily cytoplasmic , use 0.1-0.5% Triton X-100 for 5-10 minutes to ensure antibody access to intracellular compartments.
Antibody dilution: Start with a dilution range of 1:200-1:800 as recommended for most OTUB1 antibodies in IF/ICC applications , and optimize based on signal-to-noise ratio.
Signal validation: U2OS cells have been validated for positive IF/ICC detection with certain OTUB1 antibodies and can serve as a positive control.
Co-localization studies: Consider co-staining with markers for specific cellular compartments, especially if investigating interaction with binding partners or substrates.
Blocking: Use 1-5% BSA or normal serum (from the species of the secondary antibody) in PBS to reduce background.
Nuclear counterstain: Include DAPI or Hoechst to visualize nuclei, which helps distinguish OTUB1's predominantly cytoplasmic localization from any nuclear signal that might indicate specific functional states or interactions.
Imaging parameters: Use confocal microscopy for better resolution of subcellular localization, particularly when examining potential interactions with ubiquitinated substrates or E2 enzymes.
For successful immunoprecipitation (IP) of OTUB1:
Antibody selection: Choose antibodies specifically validated for IP applications . Not all OTUB1 antibodies that work for Western blot will be effective for IP.
Lysis conditions: Use mild lysis buffers (e.g., RIPA or NP-40 based) to preserve protein-protein interactions, especially when investigating OTUB1's interactions with E2 enzymes or substrate proteins.
Pre-clearing: Pre-clear lysates with protein A/G beads to reduce non-specific binding, which is particularly important when studying OTUB1's specific interactions.
Controls: Include appropriate controls:
IgG control from the same species as the OTUB1 antibody
Input sample (5-10% of the lysate used for IP)
If possible, OTUB1 knockout or knockdown samples as negative controls
Detection strategy: For co-IP experiments, consider whether to probe for OTUB1 or its interacting partners. When studying E2 enzyme interactions, remember that OTUB1 binds multiple E2s with similar affinities (Kd values ranging from 3.9–9.3 μM) .
Washing stringency: Balance between removing non-specific interactions and preserving specific ones. For studying weaker interactions, such as those with Kd values in the micromolar range, less stringent washing may be necessary.
Elution conditions: Use conditions that efficiently elute OTUB1 without denaturing potential interaction partners if co-IP is the goal.
Differentiating between OTUB1's canonical deubiquitinating activity and its non-canonical inhibitory effects on E2 enzymes requires specific experimental approaches:
Catalytic mutants: Utilize the OTUB1 C91S catalytic mutant, which lacks deubiquitinating activity but retains non-canonical functions . Comparing wild-type OTUB1 with this mutant allows you to determine whether observed effects depend on catalytic activity.
Substrate specificity analysis: OTUB1 canonically cleaves K48-linked polyubiquitin chains but not K63-linked chains . Analyze the effects on different ubiquitin linkage types to distinguish mechanism types.
E2 binding studies: For investigating non-canonical activity, determine which E2 enzymes are present in your system. OTUB1 interacts with several E2s including UBE2D1-3, UBE2E1-3, and UBE2N/UBC13 , but with different functional outcomes.
In vitro ubiquitination assays: Set up parallel reactions with recombinant components:
One set with wild-type OTUB1 to observe both activities
One set with OTUB1 C91S to isolate non-canonical effects
One set with an E2-binding deficient OTUB1 mutant to isolate canonical activity
Kinetic measurements: The canonical activity of OTUB1 alone has a high KM (approximately 85-102 μM for K48 diubiquitin) , whereas E2 binding increases OTUB1's affinity for its substrate. Measuring these parameters can help distinguish which mechanism is predominant.
Domain-specific antibodies: If available, use antibodies that specifically recognize conformational states associated with different activities of OTUB1.
To study OTUB1's functions in immune contexts:
Conditional knockout models: Utilize tissue-specific knockout models, such as CD11c-Cre OTUB1 fl/fl mice that have OTUB1-deficient dendritic cells (DCs) . These models allow for the assessment of OTUB1's role in specific immune cell populations.
Stimulation assays: Challenge OTUB1-competent and OTUB1-deficient immune cells with specific pathogen-associated molecular patterns (PAMPs) such as TLR ligands . OTUB1 promotes NF-κB activation and cytokine production in response to TLR stimulation.
Cytokine profile analysis: Measure production of key cytokines like IL-6, IL-12, and TNF in response to stimuli in the presence or absence of OTUB1 . This provides functional readouts of OTUB1's impact on immune signaling.
Mechanistic pathway dissection: Examine K48-linked deubiquitination and stabilization of the E2-conjugating enzyme UBC13, which results in increased K63-linked ubiquitination of IRAK1 and TRAF6 in the NF-κB pathway .
Infection models: Use T. gondii infection models to assess how OTUB1 in DCs affects parasite control and cytokine production . This approach allows for assessment of OTUB1's role in host defense against pathogens.
Inflammation models: Evaluate how OTUB1 affects lipopolysaccharide-induced immunopathology, where OTUB1-deficient DCs show reduced activation and cytokine production, protecting against inflammatory damage .
Single-cell analysis: Implement single-cell RNA sequencing to identify cell-specific effects of OTUB1 on immune cell activation and differentiation states.
For investigating OTUB1's interactions with E2 enzymes:
Binding affinity measurements: Use isothermal titration calorimetry (ITC) to determine equilibrium dissociation constants (Kd) between OTUB1 and E2 enzymes . This has revealed that OTUB1 binds different E2s with similar affinities (Kd values between 3.9–9.3 μM).
Functional assays: Implement ubiquitination inhibition assays to assess OTUB1's ability to non-canonically inhibit different E2s . Typical assays include:
E2 (2 μM) + E3 ligase (e.g., RNF4, 2 μM) + ubiquitin (50 μM) + varying concentrations of OTUB1 C91S
Reactions initiated with ATP and E1, then analyzed by SDS-PAGE and Western blotting
Stimulation of OTUB1 DUB activity: Measure how different E2 enzymes affect OTUB1's deubiquitinating activity using K48-linked di-ubiquitin substrates . E2 binding increases OTUB1's affinity for its substrate.
Structural studies: Use X-ray crystallography or cryo-EM to visualize the interactions between OTUB1 and different E2 enzymes, which can reveal the molecular basis for functional differences.
Domain mapping: Employ truncation or point mutants of both OTUB1 and E2 enzymes to map critical interaction interfaces that determine binding versus functional outcomes.
Cellular co-localization: Perform immunofluorescence or proximity ligation assays to visualize OTUB1-E2 interactions in cellular contexts, particularly during specific cellular processes like DNA damage response.
FRET-based interaction assays: Develop fluorescence resonance energy transfer assays to monitor OTUB1-E2 interactions in real-time in living cells, which can reveal dynamics not captured by static measurements.
To ensure antibody specificity and validate OTUB1 detection:
Validate with knockdown/knockout controls: Use siRNA/shRNA knockdown or CRISPR/Cas9 knockout samples as negative controls . The signal should be significantly reduced or absent in these samples.
Peptide competition assays: Pre-incubate the antibody with the immunizing peptide before application. If specific, the antibody signal should be blocked by the cognate peptide.
Cross-validation with multiple antibodies: Use antibodies from different sources or raised against different epitopes of OTUB1. Consistent detection patterns increase confidence in specificity.
Recombinant protein controls: Include purified recombinant OTUB1 as a positive control in Western blot applications to confirm the expected molecular weight.
Tissue expression profiling: Compare antibody staining patterns with known OTUB1 expression profiles across tissues. OTUB1 is widely expressed across tissues, with particular isoform distribution patterns .
Testing in multiple applications: If an antibody shows the expected pattern in multiple applications (WB, IHC, IF), this increases confidence in its specificity.
Mass spectrometry validation: For critical experiments, consider immunoprecipitating OTUB1 and confirming its identity by mass spectrometry.
Several factors can contribute to variability in OTUB1 detection:
Isoform expression: Different cell types or tissues may express different ratios of the two OTUB1 isoforms (31 kDa and 35 kDa) , resulting in varying band patterns or staining intensities.
Post-translational modifications: OTUB1 itself can be subject to post-translational modifications that may affect antibody recognition or alter its apparent molecular weight.
Sample preparation: Variations in lysis buffers, protein extraction efficiency, or protein degradation can impact OTUB1 detection. Use protease inhibitors and consistent protocols.
Fixation methods: For IHC/IF applications, different fixation methods can affect epitope accessibility. Paraformaldehyde fixation might preserve some epitopes better than methanol fixation.
Cellular stress or stimulation: OTUB1 expression or localization might change in response to cellular stresses, immune stimulation, or other experimental conditions .
Antibody lot-to-lot variation: Different production lots of the same antibody may show slight variations in specificity or sensitivity. When possible, use the same lot for comparative studies.
Technical factors: Variations in transfer efficiency (for Western blot), incubation times, washing stringency, or detection reagents can all contribute to experimental variability.
Cell density and growth conditions: OTUB1 expression or function may vary with cell confluence or growth conditions, particularly in studies of cell cycle-related processes.
Interpreting OTUB1-related ubiquitination patterns requires careful consideration:
Distinguish direct vs. indirect effects: Determine whether observed changes in ubiquitination are directly due to OTUB1's deubiquitinating activity or its non-canonical inhibition of E2 enzymes . Use the OTUB1 C91S mutant to separate these mechanisms.
Consider linkage specificity: Remember that OTUB1 preferentially cleaves K48-linked polyubiquitin chains but not K63-linked chains . Use linkage-specific antibodies to distinguish different ubiquitin chain types.
Analyze ubiquitin chain length: OTUB1 may preferentially cleave chains of certain lengths or act from the distal end. Use techniques like ubiquitin AQUA mass spectrometry to quantify chain lengths and linkage types.
Account for E2 specificity: OTUB1 interacts with specific E2 enzymes (UBE2D1-3, UBE2E1-3, UBE2N) but not others (e.g., UBE2G2, CDC34) . Consider which E2s are relevant in your experimental system.
Consider concentration dependence: Both stimulation of OTUB1 by E2 enzymes and non-canonical inhibition of E2 enzymes by OTUB1 occur at physiologically relevant concentrations . The total cellular ubiquitin concentration (20-85 μM) and the much lower concentration of K48 chains are important factors.
Evaluate substrate competition: Multiple OTUB1 substrates might compete for binding, affecting apparent deubiquitination efficiency.
Assess free ubiquitin levels: Free ubiquitin affects OTUB1's non-canonical activity, as binding to free ubiquitin enhances OTUB1's ability to inhibit E2 enzymes .
Control for catalytic efficiency: The KM of OTUB1 alone for K48 diubiquitin (85-102 μM) is much higher than the expected concentration of K48 chains in cells, suggesting that E2 binding (which lowers KM to 12-22 μM) may be necessary for effective DUB activity in vivo .
For exploring OTUB1's potential as a cancer therapeutic target:
Expression profiling: Use OTUB1 antibodies in IHC to analyze expression patterns across cancer tissue microarrays, correlating levels with patient outcomes .
Pathway analysis: Investigate OTUB1's interactions with cancer-associated signaling pathways including MAPK, ERα, EMT, RHOa, mTORC1, FOXM1, and p53 . Immunoprecipitation followed by pathway component detection can reveal critical interactions.
Target validation studies: Employ RNAi or CRISPR-based approaches to modulate OTUB1 levels, then use antibodies to confirm knockdown and analyze effects on potential downstream targets.
Resistance mechanism investigation: In treatment-resistant cancers, assess whether OTUB1 levels or activity are altered, potentially stabilizing oncogenic factors through deubiquitination.
Drug response biomarkers: Evaluate whether OTUB1 levels or post-translational modifications correlate with response to existing therapies, particularly those affecting protein degradation pathways.
Inhibitor development support: Use OTUB1 antibodies to validate target engagement and specificity of potential OTUB1 inhibitors through techniques like cellular thermal shift assays (CETSA).
Functional readouts: Develop immunoassays that detect OTUB1 activity rather than just presence, potentially by measuring substrate deubiquitination as a surrogate for inhibitor efficiency.
To differentiate OTUB1's compartment-specific functions:
Subcellular fractionation: Perform careful fractionation to separate cytoplasmic, nuclear, membrane, and organelle fractions, followed by Western blot analysis to determine OTUB1 distribution.
High-resolution imaging: Utilize super-resolution microscopy techniques with OTUB1 antibodies to precisely localize OTUB1 within specific cellular structures.
Proximity labeling: Employ BioID or APEX2 fusion constructs with OTUB1 to identify proximity interactors in different cellular compartments, validating findings with co-immunoprecipitation and OTUB1 antibodies.
Compartment-targeted OTUB1 variants: Create OTUB1 constructs with specific localization signals (nuclear, mitochondrial, ER, etc.) and use antibodies to confirm proper targeting and analyze compartment-specific functions.
Double immunofluorescence: Perform co-staining of OTUB1 with markers for specific organelles or structures to identify potential sites of action beyond its known cytoplasmic localization .
Stimulus-dependent translocation: Investigate whether specific stimuli (DNA damage, immune activation, stress) cause OTUB1 to relocalize, using time-course immunofluorescence studies.
Protease protection assays: For membrane-associated functions, determine whether OTUB1 is exposed to the cytosol or sequestered within membrane compartments through selective permeabilization and antibody accessibility.
For developing sophisticated OTUB1 activity assays:
FRET-based deubiquitination assays: Design substrates with fluorophore pairs on ubiquitin and target protein that produce FRET signal when conjugated and loss of signal upon OTUB1-mediated deubiquitination.
Cellular activity reporters: Develop cell-based reporters where OTUB1 activity results in stabilization of a fluorescent or luminescent readout protein targeted for K48-linked ubiquitin-mediated degradation.
High-throughput screening platforms: Establish microplate-based assays using purified components (OTUB1, E2 enzymes, ubiquitin substrates) to screen for inhibitors or activators of either canonical or non-canonical OTUB1 functions.
Affinity-based activity probes: Utilize activity-based probes that covalently modify active OTUB1, followed by detection with specific antibodies to measure the proportion of catalytically active enzyme.
Quantitative mass spectrometry: Implement targeted proteomics approaches to measure:
Changes in ubiquitination states of known OTUB1 substrates
OTUB1 interaction partners under different conditions
Post-translational modifications of OTUB1 that regulate its activity
Reconstituted systems: Establish in vitro systems with defined components to dissect:
How E2 concentration affects the balance between OTUB1's canonical and non-canonical activities
Competition between different substrates for OTUB1
Effects of potential inhibitors on both modes of OTUB1 function
Surface plasmon resonance (SPR) or bio-layer interferometry (BLI): Measure real-time kinetics of OTUB1 interactions with substrates, E2 enzymes, and potential inhibitors to understand the dynamics of these interactions.
| Application | Recommended Dilution Range | Optimal Incubation Conditions | Sample Preparation Notes |
|---|---|---|---|
| Western Blot | 1:500-1:3000 | Overnight at 4°C | 20-40 μg total protein per lane |
| Immunofluorescence | 1:200-1:800 | 1-2 hours at RT or overnight at 4°C | 4% PFA fixation, 0.1-0.5% Triton X-100 |
| Immunohistochemistry | 1:50-1:200 | 1-2 hours at RT | FFPE sections, antigen retrieval required |
| Immunoprecipitation | 2-5 μg per 1 mg lysate | Overnight at 4°C | Mild lysis buffers (RIPA or NP-40) |
| ELISA | Sample-dependent | 1-2 hours at RT | Validation required |
| E2 Enzyme | Binding Affinity (Kd) | OTUB1 DUB Stimulation | Non-canonical Inhibition by OTUB1 |
|---|---|---|---|
| UBE2D1 | 3.9 μM | Strong | Yes |
| UBE2D2 | 4.5 μM | Strong | Yes |
| UBE2D3 | 4.7 μM | Strong | Yes |
| UBE2N | 8.1 μM | Moderate | Yes |
| UBE2E1 | 9.3 μM | Weak | Yes |
| UBE2E2 | Not determined* | Weak | Yes |
| UBE2E3 | Not determined* | Not determined | Yes (except autoubiquitination) |
| UBE2G2 | No binding detected | No | No |
| CDC34 | No binding detected | No | No |
*Precipitation occurred during measurement attempts