SUMF2 antibodies are immunological reagents designed to specifically bind to the SUMF2 protein, a member of the formylglycine-generating enzyme (FGE) family. SUMF2 localizes to the endoplasmic reticulum (ER) and modulates the activity of sulfatases by interacting with SUMF1, another FGE family member . These antibodies enable researchers to investigate SUMF2's expression, localization, and functional roles in diseases such as asthma and cancer .
SUMF2 antibodies are validated for multiple experimental techniques:
Antigen retrieval methods (e.g., TE buffer pH 9.0 or citrate buffer pH 6.0) are recommended for IHC .
SUMF2 downregulation in ovalbumin (OVA)-induced asthmatic rats correlates with increased IL-13 expression in bronchial smooth muscle cells (BSMCs), suggesting its role in airway inflammation .
Immunohistochemical staining using SUMF2 antibodies revealed reduced SUMF2 protein levels in lung tissues and peripheral blood lymphocytes of asthmatic models, alongside IL-13 overexpression .
SUMF2 forms heterodimers with SUMF1 and binds sulfatases (e.g., IDS, SGSH), inhibiting SUMF1’s ability to activate these enzymes .
Forced homodimerization of SUMF2 disrupts its inhibitory effect on SUMF1, highlighting a stoichiometric regulatory mechanism .
Species Reactivity: Validated in human, mouse, and rat samples .
Consistency: Medium correlation between antibody staining and RNA expression data in the Human Protein Atlas .
SUMF2 (Sulfatase Modifying Factor 2) is a critical protein involved in the activation of sulfatases, which are essential enzymes that hydrolyze sulfate esters. SUMF2 plays a crucial role in the post-translational modification process that converts specific cysteine residues to Cα-formylglycine (FGly) in the active site of sulfatases. This modification is essential for sulfatase enzymatic function.
SUMF2 is primarily localized in the endoplasmic reticulum lumen and consists of 301 amino acids. It is expressed widely across human tissues including lung, heart, placenta, brain, liver, pancreas, skeletal muscle, and kidney, indicating its importance in multiple physiological processes . While SUMF2 has low catalytic activity when acting alone, it significantly enhances catalytic efficiency when forming heterodimeric complexes with SUMF1 .
SUMF2 antibodies are versatile research tools employed in multiple experimental techniques:
| Application | Description | Typical Dilutions |
|---|---|---|
| Western Blot (WB) | Detection of SUMF2 protein in cell/tissue lysates | 1:500-1:5000 |
| Immunoprecipitation (IP) | Isolation of SUMF2 from complex protein mixtures | 0.5-4.0 μg for 1-3 mg total protein |
| Immunohistochemistry (IHC) | Visualization of SUMF2 in tissue sections | 1:50-1:500 |
| Immunofluorescence (IF) | Cellular localization of SUMF2 | 1:50-1:500 |
| ELISA | Quantitative measurement of SUMF2 | 1:20000-1:80000 |
These applications allow researchers to examine SUMF2 expression patterns, protein-protein interactions, and localization in various experimental systems .
Both monoclonal and polyclonal SUMF2 antibodies are available for research, each with distinct characteristics:
Monoclonal SUMF2 Antibodies:
Recognize a single epitope on the SUMF2 protein
Offer high specificity and minimal cross-reactivity
Provide consistent lot-to-lot reproducibility
Example: SUMF2 Antibody (D-3), a mouse monoclonal IgG1 kappa antibody that detects human SUMF2
Polyclonal SUMF2 Antibodies:
Recognize multiple epitopes on the SUMF2 protein
Provide stronger signal amplification due to multiple epitope binding
May offer greater detection sensitivity in certain applications
More tolerant to minor protein denaturation or modifications
Example: Rabbit polyclonal antibodies against SUMF2 that react with human, mouse, and rat samples
The choice between monoclonal and polyclonal antibodies depends on the specific research requirements, with monoclonals preferred for highly specific detection and polyclonals for applications requiring enhanced sensitivity.
When selecting an SUMF2 antibody, researchers should evaluate several critical factors:
Target Species Reactivity: Ensure the antibody recognizes SUMF2 from your species of interest. Available SUMF2 antibodies have documented reactivity with human, mouse, and rat SUMF2 .
Application Compatibility: Verify that the antibody has been validated for your specific application (WB, IP, IHC, IF, ELISA). Some antibodies perform well in multiple applications, while others may be optimized for specific techniques .
Epitope Location: Consider whether the antibody targets specific domains of SUMF2 that may be relevant to your research question, particularly if studying protein interactions or post-translational modifications.
Antibody Format: Determine whether you need unconjugated antibodies or those conjugated to specific labels (HRP, PE, FITC, Alexa Fluor® conjugates) based on your detection system .
Validation Data: Review available validation data including Western blot images, IHC staining patterns, and specificity tests to ensure the antibody performs as expected .
Lot-to-Lot Consistency: For long-term studies, consider antibodies with documented lot-to-lot consistency to ensure reproducible results.
For optimal Western blotting with SUMF2 antibodies, follow this methodological approach:
Sample Preparation:
Extract proteins from cells/tissues using appropriate lysis buffers
Load 20-30 μg protein per lane (adjust based on SUMF2 expression levels)
Include appropriate positive controls (e.g., A431 cells show good SUMF2 expression)
Electrophoresis and Transfer:
Use 5-20% SDS-PAGE gels
Run at 70V (stacking gel)/90V (resolving gel) for 2-3 hours
Transfer to nitrocellulose membrane at 150 mA for 50-90 minutes
Blocking and Antibody Incubation:
Block with 5% non-fat milk in TBS for 1.5 hours at room temperature
Incubate with primary SUMF2 antibody at recommended dilution (typically 0.5-2 μg/mL) overnight at 4°C
Wash with TBS-0.1% Tween (3 times, 5 minutes each)
Incubate with appropriate HRP-conjugated secondary antibody (typically 1:5000 dilution) for 1-1.5 hours at room temperature
Detection:
Develop using enhanced chemiluminescent detection (ECL) kit
Expected band size for SUMF2 is approximately 34 kDa
Multiple bands may be observed due to different glycosylated forms of SUMF2
Troubleshooting Tips:
If background is high, increase blocking time or try different blocking agents
If signal is weak, increase antibody concentration or extend incubation times
For multiple bands, verify specificity with appropriate controls or knockdown experiments
For successful immunohistochemistry (IHC) with SUMF2 antibodies:
Tissue Preparation and Antigen Retrieval:
Use paraffin-embedded or frozen tissue sections (4 μm thickness recommended)
Perform heat-mediated antigen retrieval in EDTA buffer (pH 8.0) for optimal results
Alternative: citrate buffer (pH 6.0) may also be effective for some antibodies
Blocking and Antibody Application:
Block with 10% goat serum to reduce non-specific binding
Apply SUMF2 primary antibody at appropriate dilution (typically 1:50-1:500 or 2 μg/mL)
Detection System:
Use appropriate secondary antibody system (e.g., peroxidase-conjugated goat anti-rabbit IgG)
Incubate for 30 minutes at 37°C
Develop using DAB (3,3'-diaminobenzidine) as chromogen
Scoring and Interpretation:
Evaluate staining intensity (0-4 scale: 0=no staining, 4=strongest immunoreactivity)
Assess percentage of immunoreactive cells (0-4 scale: 0=no positive cells, 4≥75% positive cells)
Calculate composite score by multiplying intensity and percentage scores
Positive Control Tissues:
SUMF2 antibodies have shown positive IHC staining in:
Human breast cancer tissue
Human colon adenocarcinoma tissue
Human ovarian tumor tissue
Human placenta tissue
Verifying SUMF2 antibody specificity is crucial for experimental validity. Implement these methodological approaches:
Include Multiple Controls:
Positive controls: Use tissues/cells known to express SUMF2 (e.g., A431 cells, human skin tissue)
Negative controls: Include samples where the primary antibody is omitted
Isotype controls: Use non-specific antibodies of the same isotype to identify non-specific binding
Blocking peptide controls: Pre-incubate antibody with the immunizing peptide to confirm epitope-specific binding
Knockdown/Knockout Validation:
Perform siRNA knockdown or CRISPR/Cas9 knockout of SUMF2
Compare staining patterns between wild-type and SUMF2-depleted samples
Significant reduction in signal in depleted samples confirms specificity
Multiple Antibody Validation:
Use two or more antibodies that recognize different epitopes of SUMF2
Concordant results from different antibodies support specificity
Correlation with mRNA Expression:
Compare protein detection patterns with SUMF2 mRNA expression by RT-PCR or in situ hybridization
Consistent patterns between protein and mRNA expression support antibody specificity
Molecular Weight Verification:
Researchers working with SUMF2 antibodies may encounter several technical challenges:
For immunohistochemistry specifically, SUMF2 antibody staining has been successfully demonstrated in multiple tissue types with proper antigen retrieval and optimization .
When analyzing SUMF2 staining patterns across tissues, consider these methodological interpretation principles:
Baseline Expression Patterns:
Subcellular Localization Analysis:
SUMF2 is primarily localized in the endoplasmic reticulum lumen
Evaluate whether staining patterns match the expected subcellular localization
Variations in subcellular localization may indicate tissue-specific functions or protein interactions
Pathological vs. Normal Tissue Comparison:
SUMF2 antibodies have shown positive staining in various cancer tissues including breast cancer, colon adenocarcinoma, ovarian tumors, and squamous cell carcinoma
Compare expression patterns between normal and pathological tissues of the same origin
Quantify differences using standardized scoring systems (intensity and percentage of positive cells)
Context-Specific Expression:
Technical Considerations:
Different tissue types may require optimization of antigen retrieval methods
Fixation methods and tissue processing can affect epitope availability
Standardize your protocol for each tissue type after initial optimization
SUMF2 forms important interactions with SUMF1 and sulfatases. These interactions can be studied using SUMF2 antibodies through these methodological approaches:
Co-Immunoprecipitation (Co-IP):
Use SUMF2 antibodies to immunoprecipitate SUMF2 and associated proteins
Western blot analysis of immunoprecipitates with antibodies against suspected binding partners
This approach has successfully demonstrated SUMF2 interactions with SUMF1 and sulfatases
Protocol recommendation: Use 0.5-4.0 μg of SUMF2 antibody for 1.0-3.0 mg of total protein lysate
Proximity Ligation Assay (PLA):
Employ SUMF2 antibodies alongside antibodies against potential binding partners
PLA provides in situ visualization of protein interactions with high sensitivity
Especially useful for studying endogenous SUMF2 interactions in their native cellular context
Fluorescence Resonance Energy Transfer (FRET):
Pull-down Assays with Recombinant Proteins:
Bimolecular Fluorescence Complementation (BiFC):
Combine with SUMF2 antibody staining to validate protein-protein interactions
Useful for confirming the specificity of observed interactions
Research has shown that SUMF2 forms homodimers and heterodimers with SUMF1, and these interactions can be detected using appropriate antibodies in various experimental systems .
SUMF2 has been implicated in various disease processes, particularly in allergic inflammation and sulfatase-related disorders. Here are methodological approaches to investigate SUMF2 in disease models:
Expression Analysis in Disease Tissues:
Animal Model Studies:
In asthma research, SUMF2 expression has been studied in ovalbumin-induced rat models
Use SUMF2 antibodies to track protein expression changes during disease progression
Correlate SUMF2 levels with disease markers (e.g., IL-13 levels in asthma models)
Protocol example: For rat models, use SUMF2 polyclonal antibodies at 1:100 dilution for IHC
Cell-Specific Expression Analysis:
Combine SUMF2 antibody staining with cell-type markers for co-localization studies
Determine which cell populations alter SUMF2 expression in disease states
Particularly relevant for inflammatory conditions where multiple cell types are involved
Functional Inhibition Studies:
Use SUMF2 antibodies to block protein function in cell culture models
Evaluate downstream effects on sulfatase activities and related pathways
Assess changes in interacting partners like SUMF1 and IL-13
Therapeutic Response Monitoring:
Track changes in SUMF2 expression or localization following treatment interventions
Correlate SUMF2 levels with treatment efficacy and disease outcomes
Research has shown that SUMF2 interacts with IL-13 and inhibits its secretion in bronchial smooth muscle cells, which may be relevant to asthma pathogenesis .
The complex regulation of sulfatase activities involves SUMF2 interactions with SUMF1 and sulfatases. SUMF2 antibodies can help elucidate these mechanisms through:
Monitoring SUMF1-SUMF2 Complex Formation:
Assessing Post-Translational Modifications:
Subcellular Localization Studies:
Quantitative Analysis in Overexpression/Knockdown Systems:
In vitro Reconstitution Assays:
Combine purified components (SUMF1, SUMF2, sulfatases)
Use SUMF2 antibodies to verify protein presence and interactions
Assess functional outcomes on sulfatase activation
A key insight is that SUMF2 modulates sulfatase activities through a direct interaction with both SUMF1 and the sulfatases, forming a regulatory mechanism that can be studied using appropriate antibody-based techniques .
Recent research utilizing SUMF2 antibodies has revealed several important insights:
SUMF2 Interactions with Inflammatory Mediators:
Studies using yeast two-hybridization and antibody-based confirmation have shown that SUMF2 interacts with IL-13, a key cytokine in allergic inflammation
SUMF2 inhibits IL-13 secretion from bronchial smooth muscle cells, independent of IL-13 glycosylation
The interaction affects the levels of different forms of intracellular IL-13 (12-kDa vs. 17-kDa)
SUMF2 Expression in Asthma Models:
Regulatory Mechanisms of Sulfatase Activities:
Structure-Function Relationships:
Tissue-Specific Expression Patterns:
Several key questions about SUMF2 remain unresolved. Here are methodological approaches to address these knowledge gaps:
Question: How does SUMF2 expression change in response to cellular stress?
Experimental Design:
Subject cells to various stressors (ER stress, oxidative stress, hypoxia)
Use SUMF2 antibodies for Western blot and IF to quantify expression changes
Correlate with markers of stress response pathways
Include time-course analysis to capture dynamic changes
Question: What is the tissue-specific interactome of SUMF2?
Experimental Design:
Perform tissue-specific immunoprecipitation with SUMF2 antibodies
Combine with mass spectrometry to identify tissue-specific binding partners
Validate key interactions with co-immunoprecipitation and proximity ligation assays
Compare interactomes between normal and disease states
Question: How does SUMF2 trafficking between cellular compartments regulate its function?
Experimental Design:
Use subcellular fractionation followed by Western blotting with SUMF2 antibodies
Perform live-cell imaging with fluorescently labeled SUMF2 antibodies
Track changes in localization in response to signaling events
Correlate with functional outcomes on sulfatase activities
Question: What is the specific role of SUMF2 in inflammatory diseases beyond asthma?
Experimental Design:
Analyze SUMF2 expression in tissue samples from various inflammatory conditions
Use standardized IHC scoring methods to quantify expression
Correlate with disease severity markers and inflammatory cytokines
Perform functional studies in relevant cell culture models
Question: How do post-translational modifications affect SUMF2 function?
Experimental Design:
Combine immunoprecipitation with SUMF2 antibodies and mass spectrometry
Identify specific modification sites and types
Generate modification-specific antibodies if available
Create mutation constructs to assess functional impacts of modifications
SUMF2 antibodies show promise for several emerging research applications with potential clinical relevance:
Biomarker Development:
SUMF2 expression patterns could serve as diagnostic or prognostic biomarkers in diseases involving sulfatase dysregulation
Standardized immunohistochemical scoring of SUMF2 in patient samples may correlate with disease progression or treatment response
Multi-marker panels including SUMF2 and interacting partners might provide more comprehensive diagnostic information
Therapeutic Target Validation:
SUMF2 antibodies can help validate this protein as a potential therapeutic target
In asthma research, the interaction between SUMF2 and IL-13 suggests potential for intervention in allergic inflammation
Blocking or enhancing SUMF2 function could modulate sulfatase activities in conditions where these enzymes are dysregulated
Drug Mechanism Studies:
SUMF2 antibodies can help elucidate the mechanisms of drugs targeting sulfatase pathways
Monitor changes in SUMF2 expression, localization, or interactions in response to therapeutic interventions
Provide biomarkers for target engagement in clinical trials
Personalized Medicine Approaches:
Evaluate SUMF2 expression patterns in patient samples to guide treatment decisions
Identify patient subgroups that might benefit from therapies targeting SUMF2-related pathways
Develop companion diagnostics using SUMF2 antibodies for targeted therapies
Novel Antibody-Based Therapeutics:
Development of function-modulating antibodies targeting SUMF2
Creation of antibody-drug conjugates for targeted delivery to cells with aberrant SUMF2 expression
Engineering of bispecific antibodies targeting SUMF2 and interacting partners simultaneously
The research into SUMF2's interaction with IL-13 in asthma models represents a particularly promising direction, as stated in the literature: "The mechanism of SUMF2 against allergic inflammation requires further study" , indicating an important avenue for future therapeutic development.