S100A12 Antibody, FITC conjugated consists of:
Primary antibody: A polyclonal or monoclonal antibody raised against recombinant S100A12 protein (typically targeting amino acids 1–92 in humans) .
Fluorophore: FITC covalently linked to the antibody, enabling excitation at 495 nm and emission at 519 nm for green fluorescence detection .
The antibody binds specifically to S100A12, which is expressed predominantly in neutrophils and myeloid cells during inflammatory responses . FITC conjugation facilitates real-time tracking of S100A12 localization and secretion, particularly in tissues affected by chronic inflammation or cancer .
This antibody is utilized in:
Immunohistochemistry (IHC): Detects S100A12 in inflamed intestinal tissue (e.g., Crohn’s disease, ulcerative colitis) and tumor stroma .
Flow cytometry: Identifies S100A12-expressing immune cells in blood or tissue samples.
Western blotting: Confirms S100A12 protein expression (molecular weight ~10 kDa) .
Immunofluorescence microscopy: Visualizes extracellular S100A12 deposits near granulomas or crypt abscesses in inflammatory bowel disease (IBD) .
IBD: S100A12 is overexpressed in neutrophils infiltrating inflamed intestinal tissues. Serum levels correlate with disease activity (470 ng/mL in active Crohn’s vs. 75 ng/mL in controls) .
RAGE/NF-κB pathway: FITC-labeled antibodies help map S100A12-RAGE interactions, which drive NF-κB activation and perpetuate inflammation .
Hepatocellular carcinoma (HCC): High stromal S100A12 correlates with poor tumor differentiation () and vascular invasion, making it a prognostic marker .
| Parameter | S100A12-Low (n=114) | S100A12-High (n=25) | P-value |
|---|---|---|---|
| Tumor differentiation | 88 (I–II) | 13 (I–II) | 0.010 |
| Vascular invasion | 48 | 13 | 0.383 |
Specificity: Validated against recombinant S100A12 (Met1–Glu92), with cross-reactivity confirmed in pigs but not rodents .
Sensitivity: Detects S100A12 at concentrations as low as 1–5 ng/mL in ELISA-based assays .
Limitations: FITC’s susceptibility to photobleaching necessitates controlled imaging conditions.
S100A12 is a 12 kDa calcium-binding protein that belongs to the S100 family, containing two EF-hand calcium-binding motifs. It is primarily expressed in the cytoplasm and/or nucleus of myeloid cells, particularly neutrophils, monocytes, and activated macrophages . S100A12 functions as an important mediator in inflammatory processes through several mechanisms:
Acts as a ligand for the Receptor for Advanced Glycation End Products (RAGE), triggering cellular activation in mononuclear phagocytes, lymphocytes, and endothelial cells
Activates nuclear factor kappa B, a key transcription factor involved in inflammatory events
Exhibits antimicrobial properties, contributing to innate immune defense
Serves as an "alarmin" signal released during cell activation, injury or death
S100A12 has gained significant research interest due to its elevated levels in various inflammatory conditions including rheumatoid arthritis, psoriatic arthritis, Crohn's disease, ulcerative colitis, Kawasaki disease, asthma, and COPD . This makes it a valuable biomarker and potential therapeutic target for inflammatory diseases.
S100A12 plays multiple roles in inflammatory signaling cascades:
Upon calcium binding, S100A12 undergoes conformational changes that facilitate its interaction with target proteins
It is secreted from neutrophils following protein kinase C activation, often in response to cytokine stimulation or cell injury
Once released, it binds to RAGE on various cell types, including mast cells, resulting in histamine and cytokine release
The S100A12-RAGE interaction activates signaling pathways that culminate in NF-κB activation and subsequent production of pro-inflammatory cytokines such as TNFα, IL-1β, and IL-6
In asthma and COPD, S100A12 is expressed by eosinophils and macrophages in airways, particularly in regions where mast cells accumulate
Interestingly, while S100A12 generally promotes inflammation, transgenic mouse studies have revealed a potential anti-inflammatory role in certain contexts, particularly in airway smooth muscle regulation .
FITC-conjugated S100A12 antibodies offer several methodological advantages for research applications:
Direct detection without secondary antibodies, simplifying experimental protocols and reducing background
Compatibility with flow cytometry for quantitative analysis of S100A12-expressing cells
Suitability for immunofluorescence microscopy to visualize S100A12 localization within cells and tissues
Ability to perform multiplexing with antibodies conjugated to spectrally distinct fluorophores
Reduced cross-reactivity compared to indirect detection methods
Real-time visualization of protein dynamics in live cell imaging applications
The bright green fluorescence of FITC (excitation ~495 nm, emission ~519 nm) provides excellent visualization and quantification of S100A12 protein in various experimental systems.
Proper experimental controls are essential for generating reliable data with FITC-conjugated S100A12 antibodies:
Isotype control: A FITC-conjugated antibody of the same isotype (typically IgG1 κappa for many S100A12 monoclonal antibodies) but irrelevant specificity
Negative cell/tissue controls: Samples known to lack S100A12 expression
Positive cell/tissue controls: Neutrophils or tissues rich in neutrophils that naturally express high levels of S100A12
Blocking controls: Pre-absorption of the antibody with recombinant S100A12 protein to demonstrate binding specificity
Unstained controls: To establish autofluorescence baseline
Single-color controls: When performing multicolor experiments, for compensation and spectral overlap correction
Secondary-only controls: When using indirect immunofluorescence methods
These controls help distinguish specific signals from background, autofluorescence, and non-specific binding, ensuring accurate interpretation of experimental results.
The choice of fixation and permeabilization methods significantly impacts S100A12 detection quality:
Fixation options:
4% paraformaldehyde (10-15 minutes at room temperature) preserves cellular architecture while maintaining S100A12 antigenicity
Methanol fixation (-20°C for 10 minutes) may enhance detection of certain S100A12 epitopes
Avoid glutaraldehyde as it can reduce binding of antibodies to S100A12 through excessive cross-linking
Permeabilization approaches:
For intracellular S100A12: 0.1-0.5% Triton X-100 (5-10 minutes at room temperature)
For flow cytometry: 0.1% saponin in PBS with 0.5% BSA maintains permeabilization throughout staining
For delicate samples: 0.05% Tween-20 provides gentler permeabilization
For optimal results, researchers should empirically determine the ideal fixation and permeabilization combination for their specific experimental system and cell type.
Detecting S100A12 in neutrophils via flow cytometry requires special considerations:
Sample preparation:
Process samples immediately to prevent neutrophil activation and spontaneous S100A12 release
Use calcium-free buffers during initial processing to minimize S100A12 secretion
Include protease inhibitors to prevent protein degradation
Staining protocol:
Surface marker staining: Include CD66b for neutrophil identification
Fixation: 2% paraformaldehyde for 10 minutes at room temperature
Permeabilization: 0.1% saponin in staining buffer
FITC-S100A12 antibody concentration: Typically 1-5 μg/mL, titrate for optimal signal-to-noise ratio
Incubation: 30-45 minutes at room temperature in the dark
Instrument settings:
Set appropriate voltage for FITC channel based on unstained and single-stained controls
Use compensation controls when multiplexing with other fluorophores
Include viability dye to exclude dead cells which may bind antibodies non-specifically
Analysis considerations:
Gate first on intact cells (FSC/SSC), then on singlets, viable cells, and neutrophil population
Compare median fluorescence intensity (MFI) rather than percent positive cells
Use appropriate statistical tests for MFI comparison between experimental groups
This optimized protocol enables reliable quantification of S100A12 expression levels in neutrophil populations.
Effective multiplexing strategies for FITC-conjugated S100A12 antibodies include:
Panel design considerations:
FITC emissions overlap minimally with far-red fluorophores (APC, Alexa Fluor 647)
Avoid PE or PE-derivatives when using FITC unless using spectral cytometry
Reserve FITC for less abundant targets and brighter fluorophores for low-expression markers
Recommended marker combinations:
| Purpose | Marker Panel |
|---|---|
| Neutrophil characterization | FITC-S100A12, CD66b-APC, CD16-BV421, CD62L-PE-Cy7 |
| Monocyte subsets | FITC-S100A12, CD14-APC, CD16-BV421, HLA-DR-APC-Cy7 |
| Inflammation panel | FITC-S100A12, TNFα-PE-Cy7, IL-6-APC, CD45-BV510 |
Technical optimizations:
Perform sequential staining for complex panels (surface markers → fixation → permeabilization → intracellular markers)
Include FcR blocking reagent to reduce non-specific binding
Optimize antibody concentrations individually before combining
Apply appropriate compensation based on single-stained controls
Consider spectral unmixing approaches for complex panels
These multiplexing strategies allow comprehensive characterization of S100A12 in relation to other cellular markers and functional parameters.
Proper preparation of recombinant S100A12 is critical for antibody validation:
Expression system selection:
Purification protocol:
Refolding considerations:
Ensure proper folding by dialyzing against calcium-containing buffer
Verify functionality through calcium-binding assays
Storage recommendations:
Validation applications:
This methodical approach ensures the availability of high-quality recombinant S100A12 for comprehensive antibody validation.
Quantitative assessment of S100A12 in inflammatory models requires systematic methodological approaches:
Flow cytometry quantification:
Calculate relative expression using median fluorescence intensity ratios (sample MFI/isotype control MFI)
Develop calibration curves using beads with known quantities of fluorophore
Convert to Molecules of Equivalent Soluble Fluorochrome (MESF) for standardization across experiments
ELISA-based approaches:
Develop sandwich ELISA using selected monoclonal antibodies (e.g., S100A12-F5C6)
Create standard curves with purified recombinant S100A12
Express results as ng/mL or pg/mL based on calibration curve
Cell imaging quantification:
Employ software analysis of immunofluorescence images
Measure integrated density of FITC signal per cell
Standardize using calibration slides with known fluorescence values
In vivo models:
Monitor disease progression by comparing S100A12 levels between experimental groups (e.g., ETEC-challenged vs. healthy animals)
Correlate S100A12 levels with other inflammatory markers and clinical parameters
Consider longitudinal sampling to track changes over time
These quantitative approaches enable precise measurement of S100A12 levels, facilitating comparison between experimental conditions and disease states.
Successful imaging with FITC-conjugated S100A12 antibodies requires attention to several technical factors:
Photobleaching mitigation:
Use anti-fade mounting media containing DABCO or ProLong Gold
Minimize exposure time and light intensity during image acquisition
Consider using modern LED light sources rather than mercury lamps
Resolution optimization:
For subcellular localization, use high-NA objectives (1.3-1.4)
Consider super-resolution techniques (STED, SIM) for detailed localization studies
Employ deconvolution algorithms to improve image quality
Co-localization studies:
Pair FITC-S100A12 with markers for subcellular compartments to determine precise localization
Recommended combinations:
FITC-S100A12 + RAGE-Alexa647 for receptor-ligand interaction studies
FITC-S100A12 + NF-κB-Alexa594 for signaling pathway visualization
FITC-S100A12 + neutrophil granule markers for trafficking studies
Tissue imaging considerations:
Account for autofluorescence in elastin-rich tissues (lungs, vessels)
Implement spectral unmixing for tissues with high autofluorescence
Use appropriate antigen retrieval methods for formalin-fixed paraffin-embedded tissues
Live cell imaging approaches:
Consider using Fab fragments of FITC-conjugated antibodies for reduced steric hindrance
Minimize phototoxicity by reducing exposure time and increasing camera sensitivity
Include environmental controls (temperature, CO2, humidity) for physiological relevance
These considerations help researchers obtain high-quality imaging data for S100A12 localization and dynamics studies.
S100A12 expression exhibits distinct patterns across inflammatory conditions, which can be detected using FITC-conjugated antibodies:
Respiratory disorders:
In asthma and COPD: Predominantly expressed by neutrophils, and also by eosinophils and macrophages in regions where mast cells accumulate
S100A12 is one of the most abundant proteins in the lungs of patients with these conditions
Transgenic mice expressing human S100A12 in smooth muscle show reduced airway inflammation and hyperreactivity in allergic lung inflammation models
Gastrointestinal disorders:
Inflammatory bowel diseases (Crohn's disease, ulcerative colitis): Elevated S100A12 in intestinal tissue and serum
Expression correlates with disease activity and mucosal inflammation
Fecal S100A12 serves as a noninvasive biomarker
Rheumatological conditions:
Rheumatoid arthritis and psoriatic arthritis: High S100A12 concentrations in synovial fluid and serum
Expression primarily in infiltrating neutrophils at sites of inflammation
Levels correlate with disease activity scores
Infectious diseases:
Bacterial infections (e.g., ETEC F4ac challenge in piglets): Significant increase in serum S100A12 levels
Concentration positively correlates with severity of infection-induced symptoms
Contributes to antimicrobial defense mechanisms
Understanding these differential expression patterns helps researchers select appropriate experimental models and interpret findings in the context of specific disease mechanisms.
Proper normalization of S100A12 flow cytometry data ensures reliable and comparable results:
Relative quantification approaches:
Normalize to isotype control (Sample MFI ÷ Isotype control MFI)
Calculate fold-change relative to untreated or control samples
Use ratio of S100A12 to housekeeping protein (requires dual staining)
Absolute quantification methods:
Employ quantitative flow cytometry using calibration beads
Convert fluorescence to Molecules of Equivalent Soluble Fluorochrome (MESF)
Establish standard curves with beads containing known fluorophore quantities
Batch correction strategies:
Include universal control samples across all experimental runs
Use standardized settings preserved in instrument protocols
Apply computational batch correction algorithms when combining data from multiple experiments
Statistical considerations:
Use non-parametric tests for non-normally distributed MFI data
Include sufficient biological replicates (minimum n=3)
Report both mean/median and measures of variability (SD, SEM, or IQR)
Recommended reporting format:
| Sample Group | Median S100A12-FITC MFI | Normalized S100A12 Expression | Statistical Significance |
|---|---|---|---|
| Control | X ± SD | 1.00 | - |
| Treatment A | Y ± SD | Y/X | p-value |
| Treatment B | Z ± SD | Z/X | p-value |
These normalization strategies ensure that S100A12 expression data is robust, reproducible, and meaningfully comparable across experimental conditions.
Methodological discrepancies in S100A12 detection can be systematically addressed:
Common discrepancies and solutions:
Flow cytometry vs. ELISA discrepancies:
Flow cytometry measures cellular content while ELISA detects secreted protein
Solution: Measure both intracellular and supernatant S100A12 to capture total expression
Correlation analysis between methods helps identify systematic differences
Antibody epitope accessibility issues:
Fresh vs. frozen sample variations:
Freezing can affect S100A12 detection in some sample types
Solution: Process all samples consistently and include frozen/thawed controls
Establish correction factors if comparing fresh and stored samples
Signal quantification differences:
Fluorescence intensity measurement vs. concentration estimation
Solution: Use calibration standards across platforms when possible
Report relative changes consistently rather than absolute values when comparing methods
Validation approaches:
mRNA expression correlation using qPCR
Mass spectrometry verification of protein identity
Cross-validation using multiple antibody clones (e.g., S100A12-F5C6, S100A12-D10F10)
Reporting recommendations:
Clearly describe all methodological details in publications
Acknowledge limitations of each detection method
Present data from multiple techniques when available
Discuss potential reasons for observed discrepancies
This systematic approach helps researchers reconcile differences between detection methods and strengthen confidence in experimental findings.
Rigorous co-localization analysis of S100A12 with other proteins requires methodical approaches:
Qualitative assessment:
Visual inspection of merged channels in overlay images
Orthogonal views (XY, XZ, YZ) for 3D confirmation
Line profile analysis across regions of interest
Quantitative co-localization metrics:
Pearson's correlation coefficient (PCC): Measures linear correlation between fluorescence intensities (-1 to +1)
Manders' overlap coefficient (MOC): Proportion of S100A12 signal overlapping with second protein (0 to 1)
Intensity correlation quotient (ICQ): Determines whether intensities vary synchronously
Advanced analysis approaches:
Object-based co-localization: Identify discrete structures rather than pixels
Distance-based measurements: Calculate minimum distances between S100A12 and target structures
Super-resolution data analysis: Apply specialized algorithms for nanoscale co-localization
Recommended software tools:
| Software | Features | Best For |
|---|---|---|
| JACoP (ImageJ plugin) | PCC, MOC, intensity correlation | General co-localization analysis |
| Imaris | 3D co-localization, object-based analysis | Volumetric datasets |
| CellProfiler | High-throughput analysis, customizable pipelines | Large-scale studies |
Controls and validation:
Positive control: Co-staining known interacting proteins
Negative control: Co-staining proteins in distinct cellular compartments
Random co-localization control: Artificially randomized images
Physical validation: Proximity ligation assay or immunoprecipitation
These approaches enable researchers to reliably determine whether S100A12 physically associates with other proteins of interest, providing insights into its functional interactions and signaling pathways.
Non-specific binding issues can be systematically addressed through the following troubleshooting protocol:
Common issues and solutions:
High background fluorescence:
Increase blocking time (use 5-10% normal serum from the species unrelated to the primary antibody)
Add 0.1-0.3% Triton X-100 to blocking buffer to reduce hydrophobic interactions
Include 0.05% Tween-20 in wash buffers
For tissues, use Image-iT FX signal enhancer before antibody incubation
Fc receptor binding:
Include Fc receptor blocking reagent (10-20 μg/mL) before antibody addition
Use F(ab')2 fragments instead of whole IgG antibodies
Increase blocking serum concentration to 10%
Dead cell artifact staining:
Include viability dye in flow cytometry panels
For tissues, extend washing steps and use fresh fixatives
Remove necrotic regions from tissue sections before staining
Cross-reactivity with similar proteins:
Optimization checklist:
| Parameter | Starting Point | Optimization Range | Notes |
|---|---|---|---|
| Antibody concentration | 1:100 dilution | 1:50 - 1:500 | Titrate in 2-fold dilutions |
| Incubation time | 1 hour at RT | 30 min - overnight | Longer at 4°C, shorter at RT |
| Washing | 3 × 5 min | 3-5 × 5-15 min | Use gentle agitation |
| Blocking | 1 hour | 1-2 hours | Fresh blocking solution |
Validation controls:
Competitive inhibition: Pre-incubate antibody with excess recombinant S100A12
Secondary-only control: Omit primary antibody
Isotype control: Use FITC-conjugated irrelevant antibody of same isotype
Biological validation: Compare high-expressing (neutrophils) vs. low-expressing cells
These methodical troubleshooting approaches help researchers achieve specific S100A12 detection with minimal background interference.
The S100A12-RAGE interaction can be investigated through multiple complementary experimental approaches:
Co-localization studies:
Double immunofluorescence with FITC-S100A12 antibody and differently labeled RAGE antibody
Confocal microscopy to visualize potential co-localization
Super-resolution microscopy for nanoscale interaction analysis
Live cell imaging to capture dynamic interactions
Binding assays:
Surface Plasmon Resonance (SPR) to determine binding kinetics
Proximity Ligation Assay (PLA) to detect interactions in situ
FRET analysis using FITC-S100A12 and acceptor fluorophore-conjugated RAGE antibodies
Co-immunoprecipitation using S100A12-specific antibodies followed by RAGE detection
Functional interaction studies:
Blockade experiments using FITC-S100A12 antibodies to inhibit RAGE binding
Cell stimulation with recombinant S100A12 and measurement of downstream signaling
Calcium flux measurements upon S100A12 stimulation
Genetic approaches:
RAGE knockdown/knockout effects on S100A12 function
Site-directed mutagenesis of S100A12 calcium-binding domains to alter RAGE interaction
Expression of dominant-negative RAGE variants
Comparison of human S100A12 transgenic mice with normal controls
Physiological relevance testing:
Ex vivo stimulation of neutrophils/monocytes with recombinant S100A12
Measurement of inflammatory mediators (TNFα, IL-1β, IL-6) following S100A12 exposure
Assessment of S100A12-RAGE axis in relevant disease models
Correlation of S100A12-RAGE interaction with clinical parameters
These experimental approaches provide complementary evidence for the S100A12-RAGE functional relationship, enabling comprehensive understanding of this important inflammatory signaling pathway.
S100A12 FITC-conjugated antibodies can be effectively implemented in high-throughput screening (HTS) workflows:
Assay development considerations:
Miniaturization to 384-well format for increased throughput
Automation of staining, washing, and imaging steps
Development of robust positive and negative controls
Optimization of cell density and antibody concentration for maximum signal-to-background ratio
High-content screening approaches:
Automated microscopy to capture subcellular S100A12 localization
Multi-parameter phenotypic profiling including:
S100A12 expression level (FITC intensity)
Subcellular distribution
Co-localization with RAGE or other partners
Cellular morphology changes
Flow cytometry-based HTS:
Plate-based flow cytometry for rapid analysis
Multiplexing with viability dyes and additional markers
Bead-based standards for quantitative analysis
Automated compensation and analysis pipelines
Sample compatibility:
Primary neutrophils from human donors
Cell lines engineered to express S100A12
Patient-derived samples for personalized medicine applications
Tissue microarrays for pathology screening
Data analysis pipeline:
Automated image analysis using CellProfiler or similar software
Machine learning algorithms for complex phenotype classification
Statistical methods for hit identification (Z-score, SSMD)
Clustering approaches to identify compound mechanisms
Example HTS applications:
Screening for compounds that modulate S100A12 expression
Identification of inhibitors of S100A12-RAGE interaction
Discovery of drugs affecting S100A12 secretion
Evaluation of anti-inflammatory compounds in S100A12-dependent pathways
These approaches enable researchers to implement S100A12 detection in large-scale screening campaigns for drug discovery and molecular pathway elucidation.
FITC-conjugated S100A12 antibodies are opening new avenues for clinical research applications:
Biomarker development:
Flow cytometric assessment of neutrophil S100A12 expression as disease activity marker
Correlation with established clinical metrics in inflammatory conditions
Longitudinal monitoring of therapy response using standardized S100A12 detection
Integration into multiparameter immune profiling panels
Precision medicine approaches:
Stratification of inflammatory disease patients based on S100A12 expression patterns
Prediction of treatment response to biologics targeting inflammatory pathways
Identification of patients likely to benefit from RAGE-pathway inhibition
Monitoring of drug efficacy through changes in S100A12 levels
Novel therapeutic targets:
Screening for compounds that modulate S100A12-RAGE interaction
Evaluation of S100A12 neutralizing antibodies as therapeutics
Assessment of drugs affecting S100A12 secretion from neutrophils
Investigation of S100A12's pro-apoptotic effects on smooth muscle as therapeutic strategy
Diagnostic technology development:
Point-of-care tests for rapid S100A12 quantification
Multiplex platforms combining S100A12 with other inflammatory markers
Imaging approaches for visualizing S100A12 distribution in affected tissues
Novel sample types for non-invasive S100A12 detection
Translation to veterinary applications:
Extension of S100A12 detection methods to livestock disease models
Development of species-specific assays for comparative medicine
Monitoring inflammatory conditions in production animals
Evaluation of zoonotic disease mechanisms involving S100A12
These emerging applications highlight the expanding role of S100A12 detection in translational and clinical research, potentially leading to novel diagnostic and therapeutic approaches for inflammatory diseases.
Emerging technologies are poised to transform S100A12 research through several innovative approaches:
Advanced imaging technologies:
Expansion microscopy for enhanced visualization of S100A12 distribution
Lattice light sheet microscopy for high-speed 3D imaging of S100A12 dynamics
Correlative light and electron microscopy (CLEM) to link S100A12 localization with ultrastructure
Light sheet microscopy for whole-tissue S100A12 mapping
Single-cell technologies:
Single-cell proteomics to measure S100A12 alongside hundreds of other proteins
CITE-seq for combined transcriptome and S100A12 protein detection
Mass cytometry (CyTOF) with metal-conjugated S100A12 antibodies for high-parameter analysis
Spatial transcriptomics to correlate S100A12 protein with gene expression patterns
Biosensor developments:
FRET-based S100A12 activity sensors
Nanobody-based detection systems for improved tissue penetration
Aptamer-based S100A12 detection methods
Label-free detection systems using plasmonic materials
Computational advances:
Deep learning algorithms for automated quantification of S100A12 staining patterns
Integrative multi-omics approaches combining S100A12 data with transcriptomics and metabolomics
Pathway modeling of S100A12-RAGE signaling dynamics
Systems biology approaches to position S100A12 within inflammatory networks
Novel reagent development:
Small, high-affinity binders (nanobodies, affimers) to S100A12
Bispecific antibodies targeting S100A12 and related proteins simultaneously
Photoswitchable fluorescent conjugates for super-resolution imaging
Engineered antibody fragments with enhanced tissue penetration
These technological innovations promise to expand the scope and resolution of S100A12 research, enabling deeper understanding of its roles in health and disease.