S100A12 Antibody,Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism

S100A12 antibody, biotin conjugated is a monoclonal or polyclonal antibody chemically linked to biotin, enabling its use in streptavidin-based detection systems. S100A12, also known as EN-RAGE (extracellular newly identified RAGE-binding protein), is a calcium-binding alarmin secreted primarily by neutrophils and monocytes during inflammatory responses . The biotin tag facilitates signal amplification, improving the antibody's utility in low-abundance protein detection .

Applications in Research

Biotinylated S100A12 antibodies are pivotal in:

  • Inflammatory Disease Studies: Detecting S100A12 in serum and tissue samples from patients with Behçet’s disease, periodontitis, and Kawasaki disease . For example, serum S100A12 levels in active Behçet’s disease patients averaged 1,134 ng/mL, dropping to 299 ng/mL post-treatment .

  • Mechanistic Insights: Mapping S100A12-RAGE interactions in endothelial cells and monocytes to study NF-κB pathway activation .

  • 3D Tissue Models: Quantifying S100A12 secretion in inflamed oral tissue cultures, where levels increased by 3.5-fold under LPS/IFN-γ stimulation .

Periodontitis Pathogenesis

  • Saliva Biomarker: Patients with severe periodontitis exhibited 2.1-fold higher salivary S100A12 levels compared to controls, correlating with clinical disease severity .

  • Monocyte Contribution: Circulating monocytes from periodontitis patients showed elevated S100A12 expression (1.92-fold higher in intermediate monocytes vs. controls) .

Inflammatory Signaling

  • RAGE Activation: Biotinylated antibodies confirmed S100A12 binding to RAGE, inducing NF-κB-driven cytokine production (e.g., TNF-α, IL-6) in monocytes .

  • Therapeutic Targeting: Anti-S100A12 antibodies reduced inflammation in murine models of colitis, highlighting their therapeutic potential .

Supplier Comparison

Biotinylated S100A12 antibodies are available from multiple vendors:

SupplierCatalog NumberHostPrice Range
R&D Systems BAF1052Goat$529 (50 µg)
Novus Biologicals NBP2-71276BMouse€601.60 (0.1 mL)
Antibodies.com A34868Rabbit$390 (50 µL)

Validation and Quality Control

  • Specificity: Validated in WB against recombinant human S100A12, showing no cross-reactivity with other S100 proteins .

  • Sensitivity: Detects S100A12 at concentrations as low as 0.1 ng/mL in ELISA .

  • Reproducibility: Consistent performance across multiple lots, with ≤15% inter-assay variability .

Future Directions

Ongoing research explores:

  • Multiplex Assays: Pairing biotinylated S100A12 antibodies with other biomarkers for comprehensive inflammation profiling.

  • Therapeutic Monitoring: Tracking S100A12 levels in clinical trials targeting RAGE or NF-κB pathways .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Components: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
CAAF1 antibody; CAGC antibody; Calcitermin antibody; Calcium-binding protein in amniotic fluid 1 antibody; Calgranulin C antibody; Calgranulin-C antibody; Calgranulin-related protein antibody; CGRP antibody; EN RAGE antibody; EN-RAGE antibody; ENRAGE antibody; Extracellular newly identified RAGE-binding protein antibody; migration inhibitory factor-related protein 6 antibody; MRP6 antibody; Neutrophil S100 protein antibody; p6 antibody; Protein S100 A12 antibody; S100 calcium binding protein A12 antibody; S100 calcium-binding protein A12 (calgranulin C) antibody; S100 calcium-binding protein A12 antibody; S100A12 antibody; S10AC_HUMAN antibody
Target Names
S100A12
Uniprot No.

Target Background

Function

S100A12 is a calcium-, zinc-, and copper-binding protein playing a significant role in regulating inflammatory processes and immune responses. Its pro-inflammatory activity encompasses leukocyte recruitment, promotion of cytokine and chemokine production, and modulation of leukocyte adhesion and migration. It functions as an alarmin or damage-associated molecular pattern (DAMP) molecule, stimulating innate immune cells through binding to the receptor for advanced glycation end products (RAGE). RAGE binding activates MAP kinase and NF-κB signaling pathways, resulting in pro-inflammatory cytokine production and upregulation of cell adhesion molecules ICAM1 and VCAM1. It acts as a monocyte and mast cell chemoattractant, stimulating mast cell degranulation and activation, which generates chemokines, histamine, and cytokines, further inducing leukocyte recruitment to inflammatory sites. S100A12 can also inhibit the activity of matrix metalloproteinases (MMPs) 2, 3, and 9 by chelating Zn2+ from their active sites. It exhibits filariacidal and filariastatic activity and possesses antifungal activity against Candida albicans, as well as activity against Escherichia coli and Pseudomonas aeruginosa, but not against Listeria monocytogenes and Staphylococcus aureus.

Gene References Into Functions

Key Research Findings on S100A12:

  1. S100A12 activates NLRP3 inflammasomes to induce MUC5AC production in airway epithelial cells. ATP induces MUC5AC production via a similar mechanism. PMID: 29906464
  2. S100A12 does not appear to induce inflammation in dental pulp; however, RAGE may be involved in pulp inflammation in males. PMID: 28834384
  3. S100A12 is a significant predictor of lung alveolar infiltration in pulmonary tuberculosis. PMID: 27539060
  4. S100A12 increases the expression of MMP-2, MMP-9, and VCAM-1 in human airway smooth muscle cells (HASMCs) via ERK1/2 signaling pathway activation, leading to HASMC injury. PMID: 28816402
  5. S100A12 binds to CD36 at the thrombospondin-1 binding site. PMID: 27734162
  6. S100A12 exhibits antimicrobial activity and its pro-inflammatory role is supported by its interaction with Toll-like receptor 4 (TLR4). PMID: 28110121
  7. Fecal S100A12 assays are relevant in the diagnosis and management of inflammatory bowel disease. PMID: 28735301
  8. S100A12's pleiotropic actions and potential clinical significance in kidney and cardiovascular diseases are reviewed. PMID: 29080693
  9. Serum S100A12 levels correlate positively with ICU length of stay and mortality after major abdominal surgery. PMID: 27689623
  10. The binding interface between S100A12 and the V domain of RAGE has been identified. PMID: 27598566
  11. S100A12 acts as a pro-inflammatory cytokine, activating dermal fibroblasts and causing dermal fibrosis. PMID: 27840235
  12. S100A12 contributes to host defense against mycobacteria in infected macrophages. PMID: 27355424
  13. S100A12 is closely associated with psoriasis disease activity and therapeutic response. PMID: 26333514
  14. S100A12 expression is downregulated in gastric cancer (GC) and correlates with poor prognosis. PMID: 26638166
  15. Serum S100A12 levels are elevated in rheumatoid arthritis and correlate with disease activity. PMID: 26767827
  16. Elevated serum S100A12 is an independent determinant of abdominal aortic calcification progression in hemodialysis patients. PMID: 26914918
  17. Chitotriosidase and S100A12 are useful in diagnosing and assessing disease activity in familial Mediterranean fever (FMF). PMID: 26339162
  18. Elevated sRAGE, HMGB1, and S100A12, with decreased esRAGE and AGEs, distinguish patients with acute respiratory distress syndrome (ARDS) from those without. PMID: 26274928
  19. Serum S100A12 levels are elevated in acute liver failure. PMID: 25825217
  20. S100A12 is a biomarker of chronic heart failure and predicts cardiovascular events. PMID: 25438075
  21. High S100A12 mRNA expression is associated with bladder transitional cell carcinoma. PMID: 25854354
  22. S100A12 is an effective marker for inflammatory diseases. PMID: 25650963
  23. High serum S100A12 expression is associated with poor response to therapy in Crohn's disease. PMID: 25625487
  24. Elevated S100A12 levels may predict perioperative complications in patients with thoracic aortic dissection. PMID: 24691129
  25. S100A12 is induced by Helicobacter pylori infection and inhibits bacterial growth. PMID: 25964473
  26. S100A12 may be involved in the damage of biliary epithelial cells and hepatocytes in primary biliary cirrhosis. PMID: 25313445
  27. Computational studies investigated the modulation of S100A12 protein structure by different ions. PMID: 24944024
  28. Resistin, S100A12, and sRAGE are involved in the pathophysiology of Kawasaki disease. PMID: 23171632
  29. S100A12 protein is significantly associated with synovitis score in rheumatoid arthritis. PMID: 25282581
  30. EN-RAGE is an inflammatory marker for future coronary heart disease (CHD). PMID: 25341801
  31. S100A12 is associated with pulmonary inflammation and acute lung injury in cardiac surgery patients. PMID: 24887223
  32. Calcitermin, an antimicrobial peptide from human airway secretions, targets Gram-negative bacteria. PMID: 11522286
  33. Serum S100A12 concentrations correlate with angiographic coronary lesion complexity. PMID: 24341566
  34. S100A12 and high-sensitivity C-reactive protein are markers of inflammation in chronic periodontitis and type 2 diabetes. PMID: 24378957
  35. Excessive S100A12 gene expression in uremic leukocytes is associated with increased serum concentration, particularly in patients with cardiovascular disease. PMID: 23921255
  36. S100A12 and human beta-defensin 2 (hBD2) correlate with the fecal microbiota, linking the intestinal innate immune response to bacterial colonization. PMID: 24307989
  37. EN-RAGE expression is increased in Takayasu's arteritis patients. PMID: 23398829
  38. High S100A12 levels are associated with the presence and severity of coronary artery disease in patients with type 2 diabetes. PMID: 23609464
  39. High S100A12 expression is associated with intestinal inflammation and relapse in inflammatory bowel disease. PMID: 23377171
  40. Plasma S100A12 level is an independent predictor for two-year all-cause mortality. PMID: 23324110
  41. S100A12 could be a novel biomarker for predicting cardiovascular events in patients with stable coronary artery disease (CAD). PMID: 22786469
  42. Fecal S100A12 levels are significantly higher in patients with severe necrotizing enterocolitis (NEC). PMID: 22796048
  43. MMP9, HMGB1, and EN-RAGE are significantly related in diabetic patients. PMID: 22549347
  44. S100A12 is up-regulated in thoracic aortic aneurysm dissection (TAAD) and may contribute to its pathogenesis. PMID: 22818064
  45. S100A12 may contribute to osteoarthritis development by upregulating MMP-13 and VEGF. PMID: 22609404
  46. While S100A12 levels are not elevated in patients with decreased kidney function, a relation to inflammatory disease markers is observed. PMID: 21822023
  47. Serum RAGE levels are reduced in juvenile rheumatoid arthritis and correlate negatively with disease activity and S100A12 levels. PMID: 21724696
  48. Elevated plasma EN-RAGE and decreased sRAGE may contribute to systemic inflammation and carotid atherosclerosis in peritoneal dialysis patients. PMID: 21906738
  49. Lung inflammation is not exacerbated by transgenic S100A12 expression in a mouse model of allergic inflammation. PMID: 21418345
  50. Plasma S100A12 protein level is strongly associated with cardiovascular disease prevalence in hemodialysis patients. PMID: 21258041
Database Links

HGNC: 10489

OMIM: 603112

KEGG: hsa:6283

STRING: 9606.ENSP00000357726

UniGene: Hs.19413

Protein Families
S-100 family
Subcellular Location
Secreted. Cytoplasm. Cytoplasm, cytoskeleton. Cell membrane; Peripheral membrane protein. Note=Predominantly localized in the cytoplasm. Upon elevation of the intracellular calcium level, translocated from the cytoplasm to the cytoskeleton and the cell membrane. Upon neutrophil activation is secreted via a microtubule-mediated, alternative pathway.
Tissue Specificity
Predominantly expressed by neutrophils, monocytes and activated macrophages. Expressed by eosinophils and macrophages in asthmatic airways in regions where mast cells accumulate. Found in high concentrations in the serum of patients suffering from various

Q&A

What is S100A12 and why is it significant in inflammatory research?

S100A12 (also known as EN-RAGE, MRP6, CAAF1, CAGC, or CGRP) is a 10.6 kDa calcium-, zinc-, and copper-binding protein that belongs to the S100 family of low molecular weight proteins. It is characterized by two calcium-binding EF-hand motifs connected by a central hinge region . S100A12 functions as an alarmin or danger-associated molecular pattern (DAMP) molecule that plays a prominent role in regulating inflammatory processes and immune responses .

The significance of S100A12 in inflammatory research stems from its pro-inflammatory activities, which include:

  • Recruitment of leukocytes to inflammatory sites

  • Promotion of cytokine and chemokine production

  • Regulation of leukocyte adhesion and migration

  • Stimulation of innate immune cells via binding to receptor for advanced glycation endproducts (RAGE)

  • Activation of MAP-kinase and NF-kappa-B signaling pathways

  • Monocyte and mast cell chemoattraction

S100A12 expression is markedly increased at sites of inflammation, making it a valuable biomarker for inflammatory conditions such as inflammatory bowel disease (IBD) and periodontitis .

How does biotin conjugation enhance S100A12 antibody detection systems?

Biotin conjugation significantly improves S100A12 antibody utility in detection systems through multiple mechanisms:

Biotin-conjugated antibodies specific for S100A12 are essential components in sandwich enzyme immunoassay techniques, where they function as detector antibodies that bind to S100A12 captured by immobilized primary antibodies . This arrangement enables:

  • Signal amplification: The high-affinity interaction between biotin and avidin/streptavidin (Kd ≈ 10^-15 M) allows for strong signal generation with minimal background

  • Versatile detection options: Biotin-conjugated S100A12 antibodies can be paired with multiple detection systems including:

    • Avidin-conjugated horseradish peroxidase (HRP)

    • Streptavidin-fluorophore conjugates

    • Streptavidin-enzyme conjugates

  • Enhanced sensitivity: In properly optimized assays, biotin-conjugated S100A12 antibodies can contribute to detection limits as low as 0.156 ng/ml

  • Multi-step staining flexibility: The biotin-avidin system allows for separation of the antibody binding step from the detection step, enabling more complex staining protocols in techniques like immunohistochemistry

What cell populations predominantly express S100A12 and how should this inform experimental design?

S100A12 exhibits a distinct expression pattern that should directly influence experimental design decisions:

S100A12 is predominantly expressed in myeloid cells, particularly:

  • Neutrophils (highest expression)

  • Monocytes (with differential expression between monocyte subsets)

  • Expression decreases during monocyte-to-macrophage differentiation

Key experimental design considerations based on this expression pattern include:

  • Cell isolation strategies: When studying S100A12, researchers should prioritize methods that preserve neutrophil populations, which are often lost in standard peripheral blood mononuclear cell (PBMC) isolation protocols

  • Timing considerations: S100A12 gene expression is higher in classical than in non-classical monocytes , and decreases during monocyte-to-macrophage maturation, making timing of cell collection critical

  • Differential expression analysis: Flow cytometry analysis reveals that:

    • S100A8 levels in monocytes are significantly lower (approximately half) than in granulocytes from non-smokers

    • S100A9 levels in granulocytes are approximately 1.8-fold higher than S100A8

    • S100A9 levels in monocytes are approximately 2.7-fold higher than S100A8

  • Population markers: Co-staining with CD15 (for neutrophils) or CD68 (for monocyte-lineage cells) helps identify S100A12-expressing populations in tissue samples

What controls are essential when using biotin-conjugated S100A12 antibodies?

Implementing appropriate controls is critical for reliable results when using biotin-conjugated S100A12 antibodies:

  • Isotype controls: Include matched isotype controls conjugated with biotin to account for non-specific binding, particularly when examining different clinical groups

  • Blocking controls:

    • Avidin and biotin blocking solutions should be employed to prevent non-specific binding, especially in tissue sections

    • Appropriate serum blocking is necessary to reduce background staining

  • Negative tissue controls:

    • Use tissues known to lack S100A12 expression

    • Include non-inflamed tissues when studying inflammatory conditions, as S100A12 expression is typically absent in tissues from patients with inactive disease

  • Specificity controls:

    • Pre-absorption of antibody with recombinant S100A12 protein

    • Western blot confirmation of antibody specificity showing a single band at approximately 10.6 kDa

  • Cross-reactivity assessment:

    • Evaluate potential cross-reactivity with other S100 family proteins, particularly S100A8 and S100A9, which often coexist with S100A12 in myeloid cells

    • Note that while commercial assays report high specificity, limitations in current technology mean cross-reactivity may still exist with some analogues

How should researchers optimize detection protocols for S100A12 in different biological samples?

Optimization strategies must be tailored to specific sample types when detecting S100A12:

Serum/Plasma Samples:

  • ELISA detection ranges should be calibrated to expected physiological concentrations (typically 0.625-40 ng/ml in healthy individuals)

  • Serial dilutions (at least three) should be performed to ensure readings fall within the linear range of detection

  • Calibration with purified S100A12 standards spanning 0.016 to 125 ng/ml is recommended

Tissue Samples (Immunohistochemistry):

  • Antigen retrieval protocol:

    • Heat-induced epitope retrieval in citrate buffer (pH 6.0)

    • Enzymatic retrieval with proteinase K for fixed tissues

    • Peroxidase blocking with 0.3% H₂O₂

  • Detection system optimization:

    • For formalin-fixed samples, use ABC (avidin-biotin complex) followed by DAB (diaminobenzidine) development

    • For immunofluorescence, consider tyramide signal amplification to enhance sensitivity

  • Co-staining considerations:

    • When co-staining with neutrophil markers, use CD15 antibodies

    • When identifying monocyte-derived cells, employ CD68 antibodies

Cell Culture Supernatants:

  • Culture conditions significantly impact S100A12 secretion:

    • Monocyte differentiation increases S100A12 secretion over time

    • Inflammatory stimuli further enhance secretion

    • Account for these dynamics when designing time-course experiments

What are effective approaches for analyzing S100A12 expression in heterogeneous tissue environments?

Analyzing S100A12 in complex tissue environments requires sophisticated approaches:

  • Multi-parameter immunofluorescence techniques:

    • Combine S100A12 staining with lineage markers (CD15, CD68)

    • Include tissue structural markers (E-cadherin, vimentin)

    • Add functional markers (cytokine receptors, activation markers)

  • Spatial distribution analysis:

    • In inflammatory bowel disease tissues, assess S100A12 expression:

      • Around granulomatous lesions in Crohn's disease

      • Within crypt abscesses in ulcerative colitis

      • In transmigrating cells crossing the epithelium

  • Extracellular vs. intracellular expression:

    • S100A12 exhibits both intracellular expression and extracellular distribution

    • Distinguish between secreted S100A12 surrounding positive cells and intracellular expression

    • Identify potential RAGE-bearing cells in infiltrates that may bind secreted S100A12

  • Quantitative image analysis:

    • Implement digital pathology approaches for quantifying:

      • Percentage of S100A12+ cells

      • Staining intensity (mean fluorescence intensity normalized to isotype controls)

      • Spatial relationships between S100A12+ cells and tissue structures

  • Flow cytometry for tissue samples:

    • Analyze distinct CD11b populations (CD11b^hi and CD11b^lo) for differential S100A12 expression

    • Compare S100A12 levels between different myeloid subpopulations

How can researchers distinguish between S100A12 and related S100 proteins in experimental systems?

Distinguishing between closely related S100 proteins requires specific methodological approaches:

  • Antibody selection considerations:

    • Choose antibodies validated against recombinant S100A8, S100A9, and S100A12

    • Confirm specificity through Western blot analysis showing distinct molecular weights

    • Use monoclonal antibodies when possible (e.g., clone OTI1D1 for S100A12)

  • Expression pattern analysis:

    • S100A12 expression is restricted primarily to granulocytes in inflammatory tissues

    • S100A8 and S100A9 show broader expression patterns

    • Flow cytometry reveals:

      • S100A9 levels in granulocytes are approximately 1.8-fold higher than S100A8

      • S100A9 levels in monocytes are approximately 2.7-fold higher than S100A8

      • S100A12 levels in granulocytes from healthy donors are significantly lower than S100A8 or S100A9

  • Multi-color flow cytometry strategy:

    • Use distinct fluorochromes for S100A8, S100A9, and S100A12

    • Include relevant lineage markers (CD15, CD14)

    • Analyze expression ratios between proteins as potential disease biomarkers

  • Functional characterization:

    • S100A12 uniquely:

      • Acts as a monocyte and mast cell chemoattractant

      • Inhibits matrix metalloproteinases (MMP2, MMP3, MMP9) by chelating Zn²⁺

      • Possesses antimicrobial activities against C. albicans, E. coli, and P. aeruginosa

What methodological approaches can address variability in S100A12 detection across patient samples?

Addressing inter-patient variability in S100A12 detection requires systematic approaches:

  • Standardized sample processing:

    • Implement consistent protocols for:

      • Blood collection tubes (EDTA vs. heparin)

      • Processing time windows

      • Centrifugation parameters

      • Storage conditions

  • Normalization strategies:

    • Normalize S100A12 levels to:

      • Total protein concentration

      • Cell counts (for cell suspensions)

      • Tissue area (for histological analysis)

      • Relevant housekeeping proteins

  • Clinical grouping considerations:

    • Stratify patients according to:

      • Disease activity (active vs. inactive IBD)

      • Environmental factors (smokers vs. non-smokers)

      • Disease subtypes (Crohn's disease vs. ulcerative colitis)

  • Technical replicate analysis:

    • Analyze technical variability through:

      • Intra-assay coefficient of variation (CV)

      • Inter-assay CV

      • Lot-to-lot antibody variation

  • Biological correlates:

    • Correlate S100A12 levels with:

      • Clinical disease activity scores

      • Other inflammatory biomarkers

      • Neutrophil counts

      • Treatment response

How can researchers effectively measure S100A12 secretion dynamics in in vitro systems?

Measuring S100A12 secretion dynamics requires specialized experimental designs:

  • Cell culture system optimization:

    • Monocyte-derived cell cultures show increased S100A12 secretion over time

    • Inflammatory stimuli further enhance secretion

    • 3D co-culture tissue models can better recapitulate in vivo conditions

  • Time-course experimental design:

    • Monitor S100A12 secretion during:

      • Monocyte-to-macrophage differentiation

      • Following inflammatory stimulation

      • Under various polarization conditions (M1 vs. M2)

  • Quantification methodology:

    • Sandwich ELISA systems with:

      • Detection ranges optimized for expected concentrations (0.625-40 ng/ml)

      • Multiple sample dilutions to ensure linearity

      • Calibration with purified S100A12 standards (0.016-125 ng/ml)

  • Concurrent cellular analysis:

    • Pair secretion analysis with:

      • Flow cytometric analysis of intracellular S100A12

      • mRNA expression analysis

      • Evaluation of secretory pathway components

  • Stimulation protocols:

    • Compare S100A12 secretion in response to:

      • Pattern recognition receptor ligands

      • Pro-inflammatory cytokines

      • Patient-derived factors

      • Disease-relevant stimuli

What are the critical parameters for optimizing sandwich ELISA protocols using biotin-conjugated S100A12 antibodies?

Optimizing sandwich ELISA protocols requires attention to several critical parameters:

  • Coating conditions for capture antibody:

    • Optimal concentration: 10 μg/well of anti-S100A12

    • Buffer composition: 0.1 M sodium carbonate buffer, pH 9.6

    • Incubation: 16 hours at 4°C

  • Blocking parameters:

    • Recommended blocker: PBS with 0.1% Tween 20 and 0.25% bovine serum albumin

    • Blocking time: one hour at 37°C

  • Sample preparation considerations:

    • Multiple dilutions (at least three) should be tested

    • Expected detection range: 0.625-40 ng/ml

    • Lower limit of detection: approximately 0.156 ng/ml

  • Detection system parameters:

    • Biotin-conjugated anti-S100A12 antibody followed by:

    • Avidin-conjugated horseradish peroxidase (HRP)

    • Substrate development optimization (timing and temperature)

  • Assay validation metrics:

    • Sensitivity: Lower limit of detection (LLD) defined as the lowest protein concentration distinguishable from zero

    • Specificity: Absence of significant cross-reactivity with analogues

    • Precision: Intra-assay and inter-assay coefficient of variation

    • Linearity: Dilution linearity across the reportable range

What flow cytometry approaches best characterize S100A12 expression in myeloid subpopulations?

Optimized flow cytometry protocols for S100A12 should incorporate:

  • Cell preparation considerations:

    • Fresh vs. fixed samples (impact on epitope recognition)

    • Permeabilization protocol optimization for intracellular S100A12 detection

    • Red blood cell lysis method selection to preserve granulocytes

  • Gating strategy design:

    • Identify distinct myeloid populations:

      • CD11b^hi vs. CD11b^lo granulocytes

      • Classical vs. non-classical monocytes (based on differential S100A12 expression)

      • Mature vs. immature neutrophils

  • Quantification approach:

    • Mean fluorescence intensity (MFI) normalized to isotype controls

    • Percent positive cells above threshold

    • Comparative ratio analysis between:

      • S100A12:S100A8 expression (significant differences between cell types)

      • S100A12:S100A9 expression

  • Panel design considerations:

    • Include markers for:

      • Neutrophil maturation (CD10, CD16)

      • Monocyte subsets (CD14, CD16)

      • Activation status (CD11b, CD66b)

      • Related S100 proteins (S100A8, S100A9)

  • Control samples:

    • Isotype controls for each fluorochrome

    • FMO (fluorescence minus one) controls

    • Biological controls (healthy vs. diseased samples)

How can immunohistochemistry protocols be optimized for S100A12 detection in tissue sections?

Immunohistochemistry protocol optimization should address:

  • Tissue preparation protocols:

    • Fixation: 4% paraformaldehyde vs. formalin (impact on epitope preservation)

    • Section thickness optimization (typically 4-5 μm)

    • Deparaffinization and rehydration steps

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval in citrate buffer

    • Enzymatic retrieval options

    • Peroxidase blocking with 0.3% H₂O₂

  • Blocking strategies:

    • Avidin and biotin blocking solutions (critical for biotin-conjugated antibodies)

    • Appropriate serum blocking to reduce background

  • Primary antibody incubation:

    • Recommended antibody: monoclonal anti-S100A12 (clone OTI1D1)

    • Incubation conditions: overnight at 4°C

    • Optimal dilution determination

  • Detection system:

    • For chromogenic detection:

      • Biotinylated secondary antibody

      • ABC complex

      • DAB solution development

    • For fluorescence:

      • Fluorophore-conjugated streptavidin

      • Signal amplification options for low-abundance targets

What data analysis approaches best quantify S100A12 expression across different experimental platforms?

Comprehensive data analysis for S100A12 should incorporate:

  • Flow cytometry data analysis:

    • Mean fluorescence intensity (MFI) normalized to isotype control

    • Percent positive cells above threshold

    • Population-specific expression (CD11b^hi vs. CD11b^lo)

  • ELISA data interpretation:

    • Standard curve fitting (4-parameter logistic regression recommended)

    • Sample dilution linearity assessment

    • Concentration calculation accounting for dilution factors

    • Quality control metrics (CV%, recovery %)

  • Immunohistochemistry quantification:

    • Digital image analysis of:

      • Staining intensity

      • Percent positive cells

      • Spatial distribution patterns (e.g., around granulomatous lesions in CD, crypt abscesses in UC)

  • Western blot analysis:

    • Densitometry normalized to:

      • Loading controls

      • Total protein stains

      • Reference standards

  • Statistical approaches:

    • Non-parametric tests for biomarker comparisons

    • Correlation analysis with clinical parameters

    • Multivariate analysis incorporating other inflammatory markers

How can S100A12 detection be integrated into studies of inflammatory diseases?

Integration of S100A12 analysis into inflammatory disease research should consider:

  • Multi-compartment analysis:

    • Tissue expression (immunohistochemistry)

    • Circulating levels (serum ELISA)

    • Local secretion (saliva, stool, or other relevant fluids)

  • Disease-specific considerations:

    • Inflammatory Bowel Disease:

      • S100A12 is strongly expressed by neutrophils infiltrating inflamed bowel tissue

      • High circulating amounts present in serum of patients with CD and UC

      • Expression patterns differ between CD (around granulomatous lesions) and UC (crypt abscesses)

    • Periodontitis:

      • Increased secretion by monocytes from periodontitis patients

      • Higher expression in gingival tissue from periodontitis patients

      • Elevated levels in saliva correlate with clinical periodontal parameters

  • Longitudinal sampling strategies:

    • Monitor S100A12 levels during:

      • Disease flares and remissions

      • Treatment responses

      • Disease progression

  • Correlation with other inflammatory markers:

    • Traditional markers (CRP, ESR)

    • Cytokine profiles

    • Cell population dynamics

What experimental approaches can elucidate S100A12 functions beyond its use as a biomarker?

Functional studies of S100A12 should incorporate:

  • Receptor-interaction studies:

    • S100A12 binds to:

      • Receptor for advanced glycation end products (RAGE)

      • TLR4

    • Binding activates MAP-kinase and NF-kappa-B signaling pathways

  • Functional consequence analysis:

    • Pro-inflammatory cytokine production

    • Cell adhesion molecule (ICAM1, VCAM1) upregulation

    • Leukocyte recruitment and migration

    • Mast cell degranulation and activation

  • Enzymatic inhibition studies:

    • S100A12 inhibits matrix metalloproteinases:

      • MMP2, MMP3, and MMP9 through Zn²⁺ chelation

      • Functional implications in tissue remodeling

  • Antimicrobial activity investigation:

    • Antifungal activity against C. albicans

    • Antibacterial activity against E. coli and P. aeruginosa

    • Mechanisms of antimicrobial action

  • Tissue model approaches:

    • 3D co-culture tissue models

    • Organoid systems

    • Ex vivo tissue culture models

What are the most common technical challenges when working with biotin-conjugated S100A12 antibodies?

Common technical challenges and solutions include:

  • High background in biotin-streptavidin systems:

    • Challenge: Endogenous biotin in tissues causing non-specific signal

    • Solutions:

      • Implement avidin-biotin blocking steps before antibody application

      • Use alternative detection systems for tissues with high endogenous biotin

      • Consider denaturing endogenous biotin with heat treatment

  • Epitope masking issues:

    • Challenge: Fixation-induced epitope masking

    • Solutions:

      • Optimize antigen retrieval protocols

      • Test multiple antibody clones targeting different epitopes

      • Consider alternative fixation methods

  • Sensitivity limitations:

    • Challenge: Low signal when detecting physiological S100A12 levels

    • Solutions:

      • Implement signal amplification techniques

      • Optimize primary antibody concentration and incubation time

      • Consider more sensitive detection substrates

  • Cross-reactivity concerns:

    • Challenge: Potential cross-reaction with S100A8 and S100A9

    • Solutions:

      • Validate antibody specificity by Western blot

      • Include appropriate controls (samples lacking S100A12)

      • Consider co-staining experiments to identify false positives

  • Variable detection across sample types:

    • Challenge: Inconsistent results between blood, tissue, and secretions

    • Solutions:

      • Develop sample-specific protocols

      • Adjust antibody concentrations for each sample type

      • Implement appropriate normalization strategies

How can researchers validate the performance of new lots of biotin-conjugated S100A12 antibodies?

Comprehensive validation of new antibody lots should include:

  • Specificity confirmation:

    • Western blot analysis showing appropriate molecular weight (10.6 kDa)

    • Competitive inhibition with recombinant S100A12

    • Testing against known positive and negative control samples

  • Sensitivity assessment:

    • Limit of detection determination

    • Signal-to-noise ratio calculation

    • Comparison to reference standard curves

  • Reproducibility evaluation:

    • Intra-assay coefficient of variation

    • Inter-assay coefficient of variation

    • Lot-to-lot comparison with previous standards

  • Application-specific validation:

    • ELISA: Standard curve linearity and recovery

    • Flow cytometry: Staining index and resolution

    • IHC/IF: Staining pattern consistency and background levels

  • Cross-platform consistency:

    • Correlation between results from different detection platforms

    • Absolute quantification comparisons where applicable

    • Method agreement analysis (Bland-Altman plots)

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.