SUMO2/3 antibodies are monoclonal or polyclonal reagents designed to recognize SUMO2 (Uniprot: P61956) and SUMO3, which share ~97% sequence homology. Key clones include 12F3, 8A2, 11G2, and 852908, each with distinct properties:
Clone 12F3 targets the peptide sequence CQIRFRFDGQPINE, conserved across species .
Clone 8A2 detects both monomeric and polymeric SUMO2/3 forms .
12F3 detects sub-nanogram levels of recombinant SUMO2 (≥0.6 ng) and shows 1,300x greater specificity for SUMO2/3 over SUMO1 .
8A2 is widely used but exhibits 8x lower sensitivity compared to 12F3 in western blot (WB) .
852908 detects SUMO2/3 at ~16–20 kDa in Jurkat and MCF-7 cell lines .
SUMO2/3 antibodies generally do not cross-react with SUMO1, but some (e.g., 2H8, 852908) show weak detection of SUMO4 due to sequence similarity .
Heat Shock Response: SUMO2/3 conjugates increase in HeLa cells after heat shock (43°C for 10 min) .
Knockdown Validation: Antibodies confirm SUMO2 depletion via shRNA, with signal reduction in WB .
12F3 enriches SUMO2/3-targeted proteins (e.g., TFII-I) more efficiently than 8A2, as shown in HeLa cell lysates .
Tandem Affinity Purification: Used in proteomic studies to identify >200 SUMO2/3-modified proteins during mitosis .
Mitotic Localization: SUMO2/3 localizes to chromosomes during metaphase in HeLa cells, visualized using 12F3 .
SUMO2 is essential for murine embryogenesis (Sumo2−/− embryos die at E10.5), while Sumo3−/− mice are viable .
Quantitative RT-PCR shows SUMO2 accounts for 75–80% of total SUMO isoforms in early embryos .
Small Ubiquitin-like Modifiers (SUMOs) are a family of small proteins that can be enzymatically attached to target proteins through a post-translational modification process called SUMOylation. Unlike ubiquitination which primarily targets proteins for degradation, SUMOylation participates in numerous cellular processes including nuclear transport, transcriptional regulation, apoptosis, and protein stability. SUMO2/3 antibodies are essential tools for studying these modification processes, allowing researchers to detect and analyze SUMOylated proteins in various experimental contexts. These antibodies enable visualization of both free SUMO2/3 proteins and their conjugated forms in cellular systems, making them indispensable for understanding the biological roles of SUMO modifications .
SUMO2 and SUMO3 are highly similar to each other (often collectively referred to as SUMO2/3) but share only about 45% sequence identity with SUMO1. While all SUMO proteins share a conserved ubiquitin-like domain and C-terminal diglycine cleavage/attachment site, they differ in their target proteins and biological functions. Proteomic studies have identified distinct but partially overlapping subsets of proteins modified by SUMO1 versus SUMO2/3. Additionally, SUMO2/3 have unique signaling properties and functions compared to SUMO1. All human SUMO proteins (SUMO1, SUMO2, SUMO3, and SUMO4) are translated as propeptides containing C-terminal prosegments, but they target different proteins and participate in distinct cellular processes despite using the same enzymatic machinery for conjugation .
SUMO2/3 proteins typically appear at approximately 12-20 kDa on Western blots. Human SUMO2, also known as Sentrin2 and SMT3B, is synthesized as a 95 amino acid propeptide with an approximate molecular weight of 11 kDa. In experimental detection, SUMO2/3 antibodies typically recognize bands at 16-20 kDa for the monomeric form, though this can vary slightly depending on the specific antibody and experimental conditions. For instance, the R&D Systems Human SUMO2/3 Antibody (A-718) detects specific bands for SUMO2/3 at approximately 16-20 kDa in lysates, while the Human/Mouse SUMO2/3/4 Antibody (AF3020) detects bands at approximately 12 kDa. Understanding these molecular weight ranges is crucial for properly identifying SUMO2/3 signals and distinguishing them from SUMOylated target proteins, which appear as higher molecular weight bands .
SUMO2/3 antibodies can detect multiple forms of these proteins, including:
Free/monomeric SUMO2/3 (typically appearing at 12-20 kDa)
Dimeric SUMO2/3
Polymeric SUMO2/3 chains
SUMOylated target proteins (appearing as higher molecular weight bands)
Importantly, different antibodies show varying preferences for these different forms. Some antibodies preferentially detect monomeric SUMO2/3 over polymeric forms (e.g., 12F3, 2H8, 852908, 8A2, and SM23/496), while others show less preference (e.g., 1E7, 3H12, EPR4602, MM093-14V22). Some antibodies poorly detect high molecular weight SUMO2 conjugates (e.g., 18H8 and EPR300). This variability should be considered when selecting an antibody for specific experimental purposes .
The specificity of SUMO2/3 antibodies varies considerably among different clones. While some antibodies show high specificity for SUMO2/3 with minimal cross-reactivity to other SUMO family members, many exhibit significant cross-reactivity, particularly with SUMO4. Research has demonstrated that several SUMO2/3 monoclonal antibodies cross-react with SUMO4, with clones 2H8 and 852908 showing particularly high cross-reactivity. Conversely, all four tested anti-SUMO4 monoclonal antibodies cross-reacted with SUMO2/3. This cross-reactivity is primarily due to the high sequence conservation between SUMO2/3 and SUMO4, especially in the C-terminal region (92% conservation). Researchers should be aware that when using these antibodies, signals might represent a combination of SUMO2/3 and SUMO4, rather than exclusively SUMO2/3 .
Selecting the appropriate SUMO2/3 antibody requires careful consideration of several factors:
Experimental application: Different antibodies perform optimally in different applications (Western blot, immunofluorescence, immunoprecipitation)
Target form: Consider whether you need to detect free SUMO2/3, conjugated forms, or both
Specificity requirements: Determine whether cross-reactivity with SUMO4 or other SUMO family members would impact your experimental interpretation
Based on published research, antibodies such as 8A2 and 12F3 show strong, specific signals for SUMO2/3, while others like EPR4602, EPR300, and SM23/496 show lower sensitivity. For applications requiring distinction between SUMO2/3 and SUMO4, avoid antibodies with known cross-reactivity (particularly 2H8 and 852908). Validation experiments using siRNA knockdown of specific SUMO family members can help confirm antibody specificity in your experimental system. Additionally, consider using complementary approaches, such as expressing tagged versions of specific SUMO proteins, to validate antibody performance .
Based on the available research data, SUMO2/3 antibodies generally cannot distinguish between SUMO2 and SUMO3 due to their high sequence similarity. Testing of various monoclonal antibodies has shown that they recognize both SUMO2 and SUMO3 with similar efficiency. When SUMO2/3 antibodies were evaluated in cell lines complemented with FLAG-SUMO2 or FLAG-SUMO3 after endogenous SUMO2/3 depletion, they showed similar detection of both FLAG-SUMO2 and FLAG-SUMO3. This inability to discriminate between these highly similar proteins means that experimental results using these antibodies reflect the combined SUMO2/3 modification rather than modification by individual SUMO family members. Researchers requiring distinction between SUMO2 and SUMO3 specifically would need to employ alternative approaches, such as mass spectrometry or expression of tagged versions of individual SUMO proteins .
For optimal Western blot detection of SUMO2/3, consider the following methodological recommendations:
Antibody dilution: Start with manufacturer-recommended dilutions, typically 0.5-1 μg/mL for monoclonal antibodies like A-718
Buffer conditions: Many SUMO2/3 antibodies perform optimally under reducing conditions
Membrane type: PVDF membranes are commonly used and recommended for SUMO2/3 detection
Secondary antibody selection: Match appropriately to the primary antibody host species (e.g., HRP-conjugated Anti-Rat IgG for Rat Anti-Human SUMO2/3 Monoclonal Antibody)
Expected signal patterns: Look for specific SUMO2/3 bands at approximately 16-20 kDa (or 12 kDa depending on the antibody), along with higher molecular weight bands representing SUMOylated proteins
Experimental evidence shows that some antibodies (like A-718) can effectively detect SUMO2/3 in lysates from various cell lines, including Jurkat human acute T cell leukemia and MCF-7 human breast cancer cells. The AF3020 antibody has successfully detected SUMO2/3/4 in K562 human chronic myelogenous leukemia and DU145 human prostate carcinoma cell lines .
To validate SUMO2/3 antibody specificity in your experimental system, implement these approaches:
siRNA knockdown: Deplete endogenous SUMO2 and SUMO3 using siRNA and confirm reduced antibody signal in Western blots
Complementation testing: In SUMO2/3-depleted cells, express tagged versions (e.g., FLAG-SUMO2 or FLAG-SUMO3) and confirm antibody detection
Recombinant protein testing: Test antibody reactivity against purified recombinant SUMO1-4 proteins to assess cross-reactivity
Peptide competition assays: Use synthetic peptides representing epitope regions to confirm binding specificity
Stress response: Assess antibody detection of increased SUMOylation in response to cellular stressors, a characteristic of functional SUMO2/3 antibodies
Research has shown that antibodies raised against SUMO2/3 typically show reduced immunoblot signal when probing lysates from SUMO2/3 siRNA-depleted cells, confirming their specificity. Additionally, testing with recombinant SUMO proteins can reveal cross-reactivity patterns, particularly with SUMO4 due to high sequence conservation in the C-terminal region .
The detection of free versus conjugated SUMO2/3 depends on several factors:
Antibody preference: Some antibodies preferentially detect monomeric SUMO2 over polymeric forms (e.g., 12F3, 2H8, 852908, 8A2, SM23/496), while others show less preference (e.g., 1E7, 3H12, EPR4602, MM093-14V22)
Exposure time: Longer exposure times during Western blot development may be needed to visualize higher molecular weight conjugates
Sample preparation: Cell lysis conditions and the presence of SUMO protease inhibitors significantly impact the preservation of SUMO conjugates
Cell type and conditions: Different cell lines show varying levels of free versus conjugated SUMO2/3
Cellular stress: Stress conditions typically increase SUMO2/3 conjugation
Research analysis of various SUMO2/3 antibodies has shown that for most antibodies, free SUMO protein typically accounts for only 10-20% of the total signal in immunoblots (for antibodies Y299, 12F3, 2H8, 852908, 8A2, ARC1382, EPR7163, JJ-085, and IOO-19). The majority of MAbs detect primarily conjugated forms. Understanding these detection biases is crucial for properly interpreting experimental results, especially when quantifying changes in SUMOylation patterns .
SUMO2/3 antibodies are valuable tools for investigating stress-induced SUMOylation, as SUMO2/3 conjugation typically increases under various stress conditions. To effectively study stress-induced SUMOylation:
Research has demonstrated substantial variability between SUMO2/3 antibodies in their ability to detect increased SUMOylation in response to various stress agents. When designing such experiments, preliminary testing with multiple antibodies may be necessary to identify those that most sensitively detect stress-induced changes in your experimental system. Additionally, combining global SUMOylation analysis with investigation of specific target proteins can provide more comprehensive insights into stress-response mechanisms .
When using SUMO2/3 antibodies for immunoprecipitation (IP) of SUMOylated proteins, researchers should consider:
Antibody selection: Not all SUMO2/3 antibodies perform equally in IP applications; testing multiple antibodies may be necessary
Denaturing conditions: Harsh denaturing conditions may be required to disrupt SUMO proteases and preserve SUMOylated proteins
Control IPs: Include isotype control antibodies to identify non-specific binding
Validation with known targets: Confirm IP efficiency using known SUMO2/3 targets (e.g., RanGAP1 or KAP1)
Complementary approaches: Consider combining with tandem affinity purification using tagged SUMO constructs
Research has shown significant variability between SUMO2/3 antibodies as enrichment reagents for SUMOylated proteins. To maximize success, preliminary testing with known SUMOylated proteins like RanGAP1 is advisable to identify antibodies with optimal IP performance in your experimental system. Additionally, be aware that some antibodies may preferentially immunoprecipitate specific subsets of SUMOylated proteins, potentially biasing your results toward certain targets .
To distinguish SUMOylation patterns across cellular compartments using SUMO2/3 antibodies:
Immunofluorescence microscopy: Use SUMO2/3 antibodies validated for immunofluorescence to visualize the subcellular distribution of SUMO2/3-modified proteins
Subcellular fractionation: Separate nuclear, cytoplasmic, and other cellular compartments before Western blot analysis with SUMO2/3 antibodies
Co-localization studies: Combine SUMO2/3 antibodies with markers for specific organelles or cellular structures
High-resolution microscopy: Consider super-resolution techniques for more detailed localization studies
Validation controls: Include SUMO2/3 knockdown controls to confirm antibody specificity in imaging applications
Different subcellular compartments often contain distinct profiles of SUMOylated proteins. For example, SUMO2/3 modifications are prominent in the nucleus, particularly in nuclear bodies and at kinetochores during cell division. CENP-E, a kinetochore-associated protein, has been found to be specifically modified by SUMO-2/3 and possesses SUMO-2/3 polymeric chain-binding activity essential for its kinetochore localization. When designing compartment-specific SUMOylation studies, select antibodies with demonstrated performance in the relevant application (Western blot of fractionated samples or immunofluorescence) and include appropriate controls to validate subcellular localization patterns .
Variations in SUMO2/3 banding patterns across different cell lines reflect biological differences in SUMOylation profiles and should be interpreted considering:
Cell-type specific SUMOylation targets: Different cell types express varied sets of proteins that can be SUMOylated
Baseline stress levels: Cell lines may have different basal stress conditions affecting SUMO2/3 conjugation
SUMO machinery expression: Variations in expression levels of SUMO conjugation and deconjugation enzymes
Free vs. conjugated SUMO ratio: The proportion of free versus conjugated SUMO2/3 varies between cell types
Sample preparation effects: Differences in lysis conditions can affect preservation of SUMO conjugates
Research analyzing SUMO2/3 patterns across multiple cell lines (A427, CAL51, CALU6, HCT116, HEK293, and U2OS) showed that while some antibodies detected primarily conjugated forms, others detected both free and conjugated SUMO2/3, with free SUMO accounting for 10-20% of the total signal. These differences represent true biological variation rather than technical artifacts. When comparing SUMOylation patterns between cell lines, it's important to use consistent sample preparation methods and to interpret differences in the context of the cell type's biology and stress status .
Common technical issues with SUMO2/3 antibodies and their solutions include:
Technical Issue | Potential Causes | Solutions |
---|---|---|
Weak signal | Insufficient antibody concentration, low SUMOylation levels | Increase antibody concentration, optimize exposure time, enrich for SUMOylated proteins |
High background | Non-specific binding, excessive antibody | Optimize blocking conditions, titrate antibody concentration, increase washing stringency |
Unexpected molecular weight bands | Cross-reactivity, degradation products, antibody preference for specific forms | Validate with SUMO2/3 knockdown, use fresh samples with protease inhibitors |
Inconsistent results | Variability in SUMO2/3 conjugation levels, stress during sample handling | Standardize sample collection and processing, control for stress conditions |
Cross-reactivity with SUMO4 | Epitope conservation between SUMO2/3 and SUMO4 | Select antibodies with validated specificity, confirm with recombinant protein controls |
Research has demonstrated that many technical issues stem from the variable performance characteristics of different SUMO2/3 antibodies. Some antibodies poorly detect high molecular weight SUMO2 conjugates (18H8 and EPR300), while others show preferences for different SUMO forms. Understanding these antibody-specific behaviors can help troubleshoot technical issues and optimize experimental conditions .
To distinguish genuine SUMO2/3 signals from artifacts:
Include knockdown controls: Perform siRNA-mediated depletion of SUMO2/3 to confirm signal specificity
Use multiple antibodies: Compare results using different SUMO2/3 antibodies with distinct epitopes
Include recombinant protein controls: Test antibody reactivity against purified SUMO proteins
Analyze expected molecular weight patterns: Genuine SUMO2/3 signals should appear at characteristic molecular weights (free SUMO at 12-20 kDa)
Stress response validation: Confirm that SUMO2/3 conjugation increases in response to cellular stressors
Research has shown that antibody validation experiments, particularly using siRNA knockdown of SUMO2/3, can effectively distinguish specific signals from artifacts. When SUMO2/3 antibodies were tested in cell lines complemented with FLAG-SUMO2 or FLAG-SUMO3 after endogenous SUMO2/3 depletion, specific antibodies showed reduced immunoblot signal in SUMO2/3-depleted samples. Additionally, peptide competition assays can help map epitope locations and understand potential cross-reactivity, allowing better discrimination between specific signals and artifacts .
SUMO2/3 antibodies are valuable tools for investigating disease-related SUMOylation patterns, particularly in:
Cancer research: Examining altered SUMOylation profiles in various cancer types
Neurodegenerative disorders: Studying SUMOylation of disease-relevant proteins like α-synuclein or tau
Cardiac pathologies: Investigating stress-induced SUMOylation in cardiac dysfunction
Inflammatory conditions: Analyzing SUMOylation of immune signaling components
Viral infections: Examining host-pathogen interactions involving SUMOylation machinery
When applying SUMO2/3 antibodies to disease research, it's critical to select antibodies validated in relevant experimental systems. For example, SUMO2/3 antibodies have been successfully used to detect SUMOylated proteins in various cancer cell lines including Jurkat human acute T cell leukemia, MCF-7 human breast cancer, K562 human chronic myelogenous leukemia, and DU145 human prostate carcinoma cells. This suggests their applicability in studying cancer-associated SUMOylation patterns. Additionally, combining SUMO2/3 detection with analysis of specific disease-relevant protein targets can provide insights into how aberrant SUMOylation contributes to pathological mechanisms .
Recent technological advances enhancing SUMO2/3 antibody performance include:
Recombinant antibody technology: Production of high-specificity recombinant antibodies with reduced batch-to-batch variation
Epitope mapping: Precise identification of antibody binding sites to understand cross-reactivity patterns
Validation standards: More rigorous validation across multiple applications and sample types
Engineered binding domains: Development of SUMO-binding domains with enhanced specificity
Combination approaches: Using antibodies alongside mass spectrometry for improved target identification
The comprehensive characterization of antibody specificity and sensitivity, as demonstrated in recent research evaluating twenty-four anti-SUMO MAbs, has significantly advanced our understanding of antibody performance characteristics. This work has revealed that antibodies like 8A2 and 12F3 show strong, specific signals for SUMO2/3, while others exhibit varying degrees of cross-reactivity. Such detailed characterization provides researchers with better information for selecting appropriate antibodies for specific applications. Furthermore, the development of antibodies validated across multiple applications (Western blot, immunofluorescence, immunoprecipitation) offers researchers more versatile tools for studying SUMOylation in diverse experimental contexts .
For accurate quantification of SUMO2/3 conjugation levels:
Include loading controls: Normalize SUMO2/3 signal to total protein or housekeeping proteins
Analyze entire lanes: Quantify the entire range of SUMOylated proteins rather than focusing on specific bands
Use appropriate controls: Include positive controls (stress-induced samples) and negative controls (SUMO2/3 knockdown)
Account for antibody preferences: Consider whether your antibody preferentially detects specific SUMO2/3 forms
Employ replicate experiments: Perform multiple biological replicates to ensure reproducibility
Research has demonstrated that different SUMO2/3 antibodies vary in their detection preferences, with some preferring monomeric SUMO2 over polymeric forms (e.g., 12F3, 2H8, 852908, 8A2, SM23/496) and others showing less preference (e.g., 1E7, 3H12, EPR4602, MM093-14V22). These antibody-specific biases should be considered when quantifying SUMOylation levels. For comprehensive analysis, consider quantifying both free SUMO2/3 (typically at 12-20 kDa) and higher molecular weight conjugates separately, as the ratio between these forms can provide insights into cellular SUMOylation dynamics .
When comparing data generated with different SUMO2/3 antibodies:
Understand epitope differences: Identify where each antibody binds on the SUMO2/3 protein
Account for form preferences: Consider whether antibodies preferentially detect monomeric or polymeric forms
Normalize to common controls: Use identical positive and negative controls across experiments
Consider cross-reactivity profiles: Factor in known cross-reactivity with other SUMO family members
Focus on relative changes: Compare relative changes rather than absolute signal intensities
Research characterizing multiple SUMO2/3 antibodies has revealed substantial differences in their detection profiles. For example, antibodies like 18H8 and EPR300 poorly detect high molecular weight SUMO2 conjugates, while others like 12F3, 2H8, and 852908 prefer monomeric forms. Given these differences, direct comparison of absolute signal intensities between antibodies is problematic. Instead, researchers should focus on relative changes in SUMOylation patterns under different experimental conditions using the same antibody. When experiments with different antibodies must be compared, using standardized recombinant SUMO2/3 protein standards across experiments can help normalize results .
To optimize sample preparation for maximal preservation of SUMO2/3 conjugates:
Rapid processing: Minimize time between sample collection and protein denaturation
Denaturing conditions: Use strong denaturing buffers (containing SDS and urea) to inactivate SUMO proteases
Protease inhibitors: Include both general protease inhibitors and specific SUMO protease inhibitors (N-ethylmaleimide)
Temperature control: Maintain samples at 4°C or on ice when not denatured
Avoid freeze-thaw cycles: Prepare single-use aliquots to prevent conjugate degradation
The preservation of SUMO conjugates is particularly challenging due to the activity of SUMO-specific proteases (SENPs) that can rapidly deconjugate SUMOylated proteins during sample preparation. Research has shown that denaturing conditions effectively inactivate these enzymes, preserving the SUMOylation state of proteins. When comparing SUMOylation patterns across different experimental conditions, consistent sample preparation is crucial to ensure that observed differences reflect biological variations rather than technical artifacts from sample handling .
Essential controls for SUMO2/3 immunofluorescence studies include:
Knockdown/knockout validation: Cells with SUMO2/3 depletion to confirm antibody specificity
Primary antibody omission: To assess background from secondary antibody
Blocking peptide competition: Using the antibody's epitope peptide to confirm binding specificity
Stress response control: Cells exposed to known SUMO-inducing stressors (positive control)
Multiple antibody validation: Using different SUMO2/3 antibodies to confirm localization patterns
Research evaluating SUMO antibodies has highlighted the importance of proper validation in immunofluorescence applications. When conducting such studies, it's critical to include appropriate controls to distinguish genuine SUMO2/3 signals from artifacts. Additionally, co-staining with markers for specific subcellular compartments can help validate the expected localization patterns of SUMO2/3, which is predominantly nuclear under normal conditions but can show altered distribution under stress or during specific cell cycle phases .
Monoclonal and polyclonal SUMO2/3 antibodies offer different advantages and limitations:
Characteristic | Monoclonal Antibodies | Polyclonal Antibodies |
---|---|---|
Specificity | Generally higher specificity to single epitope | Recognize multiple epitopes, potentially higher cross-reactivity |
Sensitivity | May have lower sensitivity depending on epitope accessibility | Often higher sensitivity due to recognition of multiple epitopes |
Batch-to-batch variation | Minimal variation between batches | Significant variation between batches |
Form detection | May preferentially detect specific SUMO forms | Generally detect broader range of SUMO forms |
Applications | Often optimized for specific applications | Often work across multiple applications |
Research comparing monoclonal antibodies (like A-718, clone 852908) with polyclonal antibodies (like AF3020) shows that both types can be effective for SUMO2/3 detection, but with different performance characteristics. Monoclonal antibodies often provide more consistent results with less background, while polyclonal antibodies may offer greater sensitivity, particularly for detecting SUMOylated proteins under native conditions. When selecting between these antibody types, researchers should consider their specific experimental requirements and the validated performance of available antibodies in their application of interest .
Research evaluating multiple SUMO2/3 antibodies has revealed substantial variability in their ability to detect stress-induced SUMOylation changes:
Antibody Clone | Stress Response Detection | Form Preference | Cross-reactivity |
---|---|---|---|
8A2 | Strong detection of stress-induced changes | Less preference between forms | Minimal cross-reactivity |
12F3 | Strong detection of stress-induced changes | Prefers monomeric SUMO2 | Minimal cross-reactivity |
2H8 | Moderate detection of changes | Prefers monomeric SUMO2 | Cross-reacts with SUMO4 |
852908 | Moderate detection of changes | Prefers monomeric SUMO2 | Cross-reacts with SUMO4 |
EPR4602 | Limited detection of changes | Less preference between forms | Minimal sensitivity |
EPR300 | Poor detection of high MW conjugates | - | Poor sensitivity |
Small Ubiquitin-Related Modifier (SUMO) proteins are a family of ubiquitin-like proteins that play a crucial role in various cellular processes through a post-translational modification known as SUMOylation. SUMOylation involves the attachment of SUMO proteins to target proteins, influencing their stability, activity, and subcellular localization. SUMO2 and SUMO3 are two closely related members of this family, often referred to collectively as SUMO2/3 due to their high sequence similarity and functional overlap.
The SUMOylation process is a multi-step enzymatic cascade that includes activation, conjugation, and ligation. It begins with the activation of SUMO proteins by the E1 activating enzyme, followed by their transfer to the E2 conjugating enzyme. Finally, the E3 ligase enzyme facilitates the attachment of SUMO proteins to specific lysine residues on target proteins. This modification can be reversed by SUMO-specific proteases, allowing for dynamic regulation of protein function.
SUMO2/3 proteins are involved in a wide range of cellular processes, including:
One notable interaction of SUMO2/3 is with the p65 subunit of NF-κB, a key regulator of immune and inflammatory responses. Research has shown that SUMO2/3 can stabilize p65 in the cytoplasm, preventing its translocation to the nucleus and subsequent activation of NF-κB target genes . This interaction has implications for various diseases, including hepatocellular carcinoma (HCC), where SUMO2/3-p65 interaction may play a role in tumorigenesis .
Mouse anti-human SUMO2/3 antibodies are monoclonal antibodies specifically designed to detect human SUMO2/3 proteins. These antibodies are widely used in research to study the expression, localization, and function of SUMO2/3 in various biological contexts. They are valuable tools for techniques such as immunohistochemistry, immunofluorescence, and co-immunoprecipitation, enabling researchers to investigate the role of SUMO2/3 in health and disease.