OTUB1 (Otubain-1) antibodies are designed to target the OTUB1 protein, a member of the OTU domain-containing deubiquitinase (DUB) family. These antibodies are widely used in research to investigate OTUB1's role in cellular processes such as:
Immune regulation, including T-cell activation and cytokine signaling .
Cancer progression through pathways like PD-L1 stabilization and ATF6 signaling .
CD8+ T Cell Activation: OTUB1 suppresses IL-15-mediated AKT activation in CD8+ T cells, acting as a checkpoint to limit excessive immune responses .
NF-κB Signaling: OTUB1 enhances dendritic cell (DC)-dependent cytokine production (e.g., IL-6, TNF) by stabilizing UBC13, a key E2 enzyme in NF-κB activation .
Viral Infection: OTUB1 stabilizes Influenza A virus NS2 protein, promoting viral replication, while its degradation by viral NS1 dampens antiviral responses .
PD-L1 Stabilization: OTUB1 deubiquitinates PD-L1, preventing its ER-associated degradation and enabling tumor immune evasion. Silencing OTUB1 enhances CD8+ T cell-mediated cytotoxicity .
Bladder Cancer: OTUB1 stabilizes ATF6 (activating transcription factor 6) via deubiquitination, driving tumor growth and the unfolded protein response (UPR) .
OTUB1 antibodies have proven indispensable in elucidating the enzyme’s dual roles in physiology and pathology. For example:
In cancer, OTUB1’s stabilization of PD-L1 and ATF6 highlights its potential as a therapeutic target .
In immunology, OTUB1’s regulation of T-cell activation and cytokine production underscores its importance in balancing immune responses .
Structural studies using these antibodies have clarified OTUB1’s catalytic and non-catalytic mechanisms, informing drug development .
The solution contains 1mg/ml of OTUB1 antibody in a buffer consisting of PBS at pH 7.4, 10% glycerol, and 0.02% sodium azide.
OTUB1 (OTU domain, ubiquitin aldehyde binding 1) is a cytoplasmic deubiquitinating enzyme belonging to the Peptidase C65 protein family. The canonical human OTUB1 protein contains 271 amino acid residues with a molecular mass of approximately 31.3 kDa. It functions as a hydrolase that specifically removes 'Lys-48'-linked conjugated ubiquitin from proteins, playing a crucial regulatory role in protein turnover by preventing degradation. OTUB1 is widely expressed across numerous tissue types and exists in up to two different isoforms. The importance of OTUB1 lies in its ability to regulate the stability of key proteins involved in essential cellular processes, including DNA repair mechanisms, making it a significant protein for understanding cellular homeostasis and disease mechanisms .
OTUB1 functions through two distinct mechanisms in the ubiquitination pathway. First, it acts as a canonical deubiquitinating enzyme (DUB) that cleaves ubiquitin chains from substrate proteins, particularly those with 'Lys-48' linkages which typically mark proteins for proteasomal degradation. Second, OTUB1 utilizes a non-canonical mechanism where it directly binds to and inhibits E2 ubiquitin-conjugating enzymes, thereby preventing the formation of ubiquitin chains on substrate proteins. This dual functionality allows OTUB1 to regulate protein stability through both active deubiquitination and prevention of ubiquitination. The interaction between OTUB1 and its target proteins, such as MSH2, occurs through specific binding domains, with OTUB1 utilizing its deubiquitylation catalytic center to mediate these interactions .
OTUB1 directly interacts with and stabilizes MSH2, a critical component of the DNA mismatch repair (MMR) system. MSH2 is an obligate subunit of two heterodimeric complexes: MutSα (MSH2-MSH6) and MutSβ (MSH2-MSH3), which are responsible for recognizing DNA mismatches and initiating repair. OTUB1 binds to the central domain of MSH2 through its deubiquitylation catalytic center, preventing MSH2 ubiquitination and subsequent proteasomal degradation. This stabilization is crucial for maintaining appropriate levels of MutSα and MutSβ complexes in the cell. When OTUB1 is depleted, enhanced MSH2 ubiquitination occurs, leading to degradation via the proteasome, which results in MMR deficiency. This deficiency manifests as a mutator phenotype and resistance to certain genotoxic agents commonly used in cancer therapy .
When selecting an OTUB1 antibody for research, several critical factors must be considered:
Antibody specificity: Verify that the antibody specifically recognizes OTUB1 and not other OTU domain-containing proteins. Review validation data including Western blots showing a single band at approximately 31.3 kDa.
Host species and clonality: Consider the experimental design when choosing between polyclonal and monoclonal antibodies. Polyclonal antibodies offer higher sensitivity by recognizing multiple epitopes, while monoclonal antibodies provide greater specificity and batch-to-batch consistency.
Application compatibility: Ensure the antibody is validated for your specific application (Western blot, immunohistochemistry, immunofluorescence, etc.). Different applications may require different antibody characteristics.
Epitope location: Consider antibodies targeting different regions of OTUB1 (N-terminal, C-terminal, or internal regions) depending on your research question, especially if studying specific isoforms or potential post-translational modifications.
Species reactivity: Verify cross-reactivity with your experimental model organism (human, mouse, rat, etc.) .
OTUB1 antibodies are employed in various research applications, with the most common being:
Western Blotting (WB): The predominant application for detecting OTUB1 protein expression levels, studying protein-protein interactions through co-immunoprecipitation, and examining OTUB1's role in various cellular contexts.
Immunohistochemistry (IHC): Used to examine the tissue and cellular distribution of OTUB1 in normal and pathological specimens, particularly important for cancer research.
Immunofluorescence (IF) and Immunocytochemistry (ICC): Applied to visualize the subcellular localization of OTUB1 and its potential co-localization with interacting proteins like MSH2.
Flow Cytometry (FACS): Used in some cases to quantify OTUB1 levels in specific cell populations.
ELISA: Employed for quantitative detection of OTUB1 in biological samples.
The specific application determines the optimal antibody format, with certain conjugated antibodies being more suitable for particular techniques. For example, unconjugated primary antibodies are typically used for Western blotting, while directly conjugated antibodies might be preferred for multi-color immunofluorescence .
Validating OTUB1 antibody specificity involves multiple complementary approaches:
Positive and negative control samples: Use cell lines or tissues known to express OTUB1 (positive control) and those with low or no expression (negative control). Additionally, OTUB1 knockout or knockdown samples provide excellent negative controls.
Multiple antibody comparison: Test several antibodies targeting different OTUB1 epitopes and compare their detection patterns. Consistent results across antibodies increase confidence in specificity.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application. This should block specific binding and eliminate the true OTUB1 signal.
Overexpression verification: Analyze samples overexpressing tagged OTUB1 (e.g., FLAG-tagged or HA-tagged) and confirm detection with both the OTUB1 antibody and tag-specific antibodies.
Western blot analysis: Verify that the antibody detects a single band at the expected molecular weight (approximately 31.3 kDa for the canonical human OTUB1).
Cross-reactivity testing: If the antibody is intended for use in multiple species, verify specificity across all target species. This is particularly important as sequence homology varies between species .
OTUB1 regulates MSH2 stability through its interaction with the central domain of MSH2, employing both canonical and non-canonical deubiquitinating mechanisms. Through its canonical activity, OTUB1 removes ubiquitin chains from MSH2, preventing its degradation via the proteasome. More significantly, OTUB1 utilizes its non-canonical function to inhibit E2 ubiquitin-conjugating enzymes, thereby preventing the ubiquitination of MSH2 in the first place.
When OTUB1 is depleted or its function is impaired, particularly its non-canonical E2-inhibiting activity, MSH2 ubiquitination increases substantially, leading to enhanced proteasomal degradation. This degradation reduces cellular levels of MSH2 and consequently disrupts the formation and function of the MutSα (MSH2-MSH6) and MutSβ (MSH2-MSH3) complexes, which are essential for recognizing DNA mismatches.
The resulting mismatch repair deficiency manifests as a mutator phenotype, characterized by increased mutation rates and microsatellite instability. Additionally, cells with OTUB1 deficiency exhibit resistance to certain genotoxic agents, including cisplatin and MNNG, as these agents rely partially on functional MMR for their cytotoxicity. Restoring OTUB1 expression in OTUB1-knockdown cells reverses these phenotypes, confirming OTUB1's specific role in regulating MMR efficiency through MSH2 stabilization .
Studying OTUB1-MSH2 interactions requires multiple complementary experimental approaches:
Co-immunoprecipitation (Co-IP): This technique is fundamental for demonstrating direct protein-protein interactions. Both tagged protein overexpression systems (e.g., Flag-tagged MSH2 and HA-tagged OTUB1 in HEK293T cells) and endogenous protein interactions can be analyzed. Reciprocal pulldowns (using antibodies against each protein) strengthen the evidence for specific interaction.
Domain mapping: To identify specific interaction domains, truncated protein constructs are created and tested for binding. For example, studies have shown that OTUB1 interacts with the central domain of MSH2, informing our understanding of the structural basis of this interaction.
Ubiquitination assays: Cell-based ubiquitination assays involve treating cells with proteasome inhibitors (e.g., MG132) to prevent degradation of ubiquitinated proteins, followed by immunoprecipitation of the target protein (MSH2) and detection of ubiquitin conjugates. This approach reveals how OTUB1 affects MSH2 ubiquitination levels.
Functional rescue experiments: Knockdown of OTUB1 followed by re-expression of wild-type or mutant OTUB1 variants helps determine which OTUB1 functions are critical for MSH2 stabilization.
Microscopy-based co-localization: Immunofluorescence microscopy can visualize potential co-localization of OTUB1 and MSH2 within cellular compartments.
Functional assays: Measuring DNA repair efficiency, mutation rates, and sensitivity to genotoxic agents in cells with altered OTUB1 expression provides functional evidence of the significance of OTUB1-MSH2 interactions .
OTUB1 expression significantly influences cellular responses to genotoxic agents through its stabilization of MSH2 and consequent maintenance of mismatch repair (MMR) functionality. Research has revealed several critical aspects of this relationship:
Differential drug sensitivity: OTUB1-knockdown cells show increased resistance to specific genotoxic agents including cisplatin and MNNG. This resistance occurs because these agents partially rely on functional MMR for their cytotoxicity, as MMR recognizes and processes drug-induced DNA damage, triggering apoptotic pathways.
Reduced apoptotic response: Cells with depleted OTUB1 show reduced levels of cleaved PARP1 (a hallmark of apoptosis) following treatment with genotoxic agents, indicating diminished apoptotic cell death.
Reversible phenotype: Restoring OTUB1 expression in knockdown cells resensitizes them to genotoxic agents, confirming the specificity of this mechanism.
Potential biomarker role: The relationship between OTUB1 expression and drug sensitivity suggests OTUB1 could serve as a biomarker for predicting tumor response to certain chemotherapeutic agents.
Therapeutic implications: Understanding OTUB1's role provides rationale for combination therapies that might overcome resistance in tumors with low OTUB1 expression or for developing strategies to stabilize MSH2 through alternative mechanisms.
This knowledge has significant implications for personalized cancer medicine, suggesting that assessment of OTUB1 expression might help guide treatment decisions, particularly for therapies utilizing DNA-damaging agents .
When using OTUB1 antibodies in Western blotting, a comprehensive set of controls should be included to ensure reliable and interpretable results:
Positive control: Include a sample known to express OTUB1, such as HeLa or HEK293T cells, which have been documented to express detectable levels of endogenous OTUB1.
Negative control: Where possible, include OTUB1 knockdown or knockout samples to confirm antibody specificity.
Loading control: Use antibodies against housekeeping proteins (e.g., GAPDH, β-actin, or tubulin) to normalize for total protein loading differences between samples.
Molecular weight marker: Include a protein ladder to confirm that the detected band appears at the expected molecular weight (approximately 31.3 kDa for canonical OTUB1).
Antibody specificity control: Consider running a peptide competition assay where the antibody is pre-incubated with the immunizing peptide, which should eliminate specific binding.
Recombinant protein control: Include purified recombinant OTUB1 protein as a definitive positive control, particularly when testing a new antibody.
Multiple antibody validation: If possible, confirm key findings with a second OTUB1 antibody that recognizes a different epitope.
Overexpression control: For studies involving exogenous OTUB1 expression, include both non-transfected cells and cells expressing an empty vector as controls .
Optimizing OTUB1 antibodies for immunohistochemistry (IHC) requires methodical protocol development:
Antigen retrieval optimization: Test multiple antigen retrieval methods, including heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) and EDTA buffer (pH 9.0), as well as enzymatic retrieval with proteinase K. OTUB1 epitopes may be differentially masked by fixation, requiring specific retrieval conditions.
Antibody dilution titration: Test a range of antibody dilutions (typically starting with manufacturer recommendations and then testing 2-fold dilutions above and below) to determine the optimal concentration that maximizes specific signal while minimizing background.
Incubation conditions: Optimize both temperature (4°C, room temperature, or 37°C) and duration (1 hour to overnight) of primary antibody incubation.
Detection system selection: Compare different detection systems (e.g., polymer-based systems, avidin-biotin complexes) to identify the most sensitive and specific method for OTUB1 detection.
Counterstaining adjustment: Optimize hematoxylin counterstaining to provide adequate nuclear contrast without obscuring OTUB1 signal.
Positive and negative tissue controls: Include tissues known to express OTUB1 (positive control) and tissues with minimal expression (negative control). Additionally, perform technical negative controls by omitting the primary antibody.
Block optimization: Test different blocking reagents (e.g., serum, BSA, commercial blocking solutions) to minimize non-specific binding.
Validation with orthogonal methods: Confirm IHC findings using alternative techniques such as Western blotting or RNA expression analysis in the same tissues .
To effectively study OTUB1's role in protein deubiquitination, several specialized methods should be employed:
In vivo ubiquitination assays: Transfect cells with HA-tagged ubiquitin along with the substrate protein of interest (e.g., MSH2). Treat cells with proteasome inhibitors (e.g., MG132) to prevent degradation of ubiquitinated proteins. Immunoprecipitate the substrate protein and detect ubiquitin conjugates using anti-HA antibodies via Western blotting. Compare ubiquitination levels between control cells and cells with manipulated OTUB1 expression or activity.
OTUB1 activity manipulation: Use multiple approaches to alter OTUB1 function:
siRNA or shRNA-mediated knockdown
CRISPR/Cas9-mediated knockout
Overexpression of wild-type OTUB1
Expression of catalytically inactive OTUB1 mutants (e.g., C91S)
Expression of mutants defective in non-canonical E2 inhibition
Protein stability assays: Measure protein half-life using cycloheximide chase assays, comparing the degradation rate of substrate proteins in the presence or absence of functional OTUB1.
In vitro deubiquitination assays: Purify recombinant OTUB1 and ubiquitinated substrate proteins to test direct deubiquitination activity in controlled conditions.
Domain mapping: Create truncated versions of both OTUB1 and the substrate protein to identify the specific domains involved in their interaction and in substrate recognition.
Mass spectrometry-based approaches: Use quantitative proteomics to identify changes in the ubiquitinome upon OTUB1 manipulation, potentially revealing new OTUB1 substrates.
Co-localization studies: Employ immunofluorescence microscopy to visualize OTUB1 and its substrates, potentially capturing dynamic interactions during cellular responses to various stimuli .
When working with OTUB1 antibodies, researchers commonly encounter several issues that can be systematically addressed:
Weak or no signal in Western blots:
Solution: Increase protein loading (50-100 μg total protein)
Decrease antibody dilution (use more concentrated antibody)
Extend exposure time during imaging
Try enhanced chemiluminescence (ECL) substrates with greater sensitivity
Ensure transfer efficiency by staining the membrane with Ponceau S
Test different blocking agents (milk vs. BSA)
Multiple bands or non-specific binding:
Solution: Increase stringency of washing steps (more washes, higher salt concentration)
Optimize blocking conditions (try 5% BSA instead of milk, or vice versa)
Increase antibody dilution
Pre-adsorb the antibody with cell lysate from OTUB1-knockout cells
Consider using a different antibody targeting a different epitope
High background in immunohistochemistry/immunofluorescence:
Solution: Extend blocking time
Use more stringent washing
Dilute the primary antibody further
Try different blocking reagents
Optimize antigen retrieval conditions
Pre-adsorb the antibody with control tissue
Inconsistent results between experiments:
Solution: Standardize protein extraction methods
Use consistent cell culture conditions
Implement a more detailed protocol with precise timing
Prepare fresh working solutions for each experiment
Store antibodies according to manufacturer recommendations
Consider aliquoting antibodies to avoid freeze-thaw cycles .
Differentiating between OTUB1 and other OTU family proteins requires careful experimental design and specific techniques:
Antibody selection: Choose antibodies raised against unique epitopes that don't share homology with other OTU family members. Review the immunogen sequence and compare it to other OTU proteins using sequence alignment tools.
Validation using overexpression systems: Express tagged versions of OTUB1 and closely related OTU proteins (especially OTUB2) in parallel. Test whether the OTUB1 antibody specifically recognizes only OTUB1 and not the other expressed OTU proteins.
Knockdown/knockout controls: Use siRNA/shRNA against OTUB1 or CRISPR/Cas9-mediated knockout cells to confirm that the signal disappears when OTUB1 is depleted.
Mass spectrometry validation: For critical experiments, confirm antibody-based identification using mass spectrometry, which can distinguish between different OTU family members based on unique peptide sequences.
Western blot analysis: OTUB1 and other OTU family members often have different molecular weights. OTUB1 is approximately 31.3 kDa, while OTUB2 is around 27 kDa, allowing for differentiation on Western blots.
Expression pattern analysis: Different OTU family members often have distinct tissue or cellular expression patterns. Compare your results with known expression profiles to support your identification.
Functional assays: OTUB1 has a preference for cleaving K48-linked ubiquitin chains, while other OTU family members may have different specificities. Design functional assays that capitalize on these differences .
Accurately interpreting OTUB1 expression changes in the context of DNA repair deficiencies requires careful consideration of multiple factors:
Establish baselines: Determine normal OTUB1 expression levels in relevant control tissues or cell lines before comparing to experimental conditions. Consider analyzing public databases for typical expression ranges.
Multi-protein analysis: Always assess OTUB1 expression alongside its key substrates, particularly MSH2 and other MMR proteins. The functional significance of OTUB1 changes depends on corresponding changes in these interacting partners.
Functional validation: Supplement expression data with functional assays of MMR efficiency, such as:
Microsatellite instability (MSI) analysis
Mutation rate measurements using reporter systems
DNA damage response assays following exposure to MMR-dependent genotoxic agents
Quantification of MMR complex formation using co-immunoprecipitation
Causality testing: When OTUB1 expression changes are observed, test whether they are causal or consequential by:
Restoring OTUB1 to normal levels and measuring impact on MMR function
Using OTUB1 mutants that separate its different functions (catalytic activity vs. E2 inhibition)
Context consideration: Interpret OTUB1 changes in light of:
Cell cycle phase (as MMR activity and protein levels fluctuate through the cell cycle)
DNA damage status
Cellular stress conditions
Tissue type (as baseline expression and importance may vary)
Protein stability vs. expression: Distinguish between changes in OTUB1 protein level due to altered gene expression versus altered protein stability by comparing mRNA and protein levels .
Ubiquitin Aldehyde Binding 1 (OTUB1) is a protein encoded by the OTUB1 gene. It belongs to the OTU (ovarian tumor) superfamily of predicted cysteine proteases. OTUB1 is a highly specific ubiquitin iso-peptidase, which means it can cleave ubiquitin from branched poly-ubiquitin chains but not from ubiquitinated substrates .
OTUB1 is known for its role in deubiquitination, a process that removes ubiquitin from proteins. This action is crucial for regulating protein turnover by preventing degradation. OTUB1 specifically removes ‘Lys-48’-linked conjugated ubiquitin from proteins . This regulatory role is essential in various cellular processes, including protein degradation, signal transduction, and DNA repair.
OTUB1 functions by binding to free ubiquitin, which acts as an allosteric regulator. This binding increases OTUB1’s affinity for the ubiquitin-conjugating enzyme UBE2N/UBC13 and disrupts its interaction with UBE2V1 . The OTUB1-UBE2N/UBC13-free ubiquitin complex adopts a configuration that mimics a cleaved ‘Lys48’-linked di-ubiquitin chain .
OTUB1 plays a significant role in the immune system by regulating T-cell anergy. T-cell anergy is a state where T-cells become unresponsive to antigen rechallenge and no longer respond to their cognate antigen. OTUB1 achieves this through its interaction with RNF128/GRAIL, a crucial inducer of CD4 T-cell anergy .
The mouse anti-human OTUB1 antibody is a monoclonal antibody that targets the OTUB1 protein. It is used in various research applications, including Western blotting, immunohistochemistry, and immunofluorescence . This antibody helps in studying the expression and localization of OTUB1 in different tissues and cell lines.