The F13A1 monoclonal antibody is produced via hybridoma technology, where mice are immunized with synthetic peptides or recombinant fragments of human F13A1 (aa46–181 or aa1–200) . Key characteristics include:
Molecular Weight Recognition: Binds specifically to the 83 kDa F13A1 protein .
Specificity: Confirmed via HuProt™ protein arrays (19,000+ human proteins), with S-scores ≥2.5 indicating high target specificity .
Tumor Differentiation:
Vascular Diseases: F13A1 inhibition reduces angiogenesis by downregulating thrombospondin-1, offering potential in treating venous thromboembolism and systemic sclerosis .
Cancer Metastasis: Inflammatory monocytes expressing F13A1 facilitate fibrin cross-linking, promoting lung squamous carcinoma invasion .
Neurological Roles: Linked to amyloid-β deposition in Alzheimer’s disease and cerebral amyloid angiopathy .
Stability: Stable at -20°C for one year; avoid freeze-thaw cycles .
Formats: Available unconjugated or conjugated to CF® dyes (e.g., CF405M, CF640R) .
The F13A1 monoclonal antibody was developed using immunization and hybridoma technology. To produce the antibody, B cells were extracted from the spleen of mice previously immunized with a synthesized peptide derived from human F13A1. These B cells were then fused with myeloma cells to create hybridomas. Following screening and selection, the F13A1-generating hybridomas were cultured in the mouse abdominal cavity. The F13A1 monoclonal antibody was purified from mouse ascites using affinity chromatography with specific immunogens. This purified F13A1 monoclonal antibody has been validated for use in ELISA and IHC applications.
F13A1 plays a crucial role in the blood clotting process. It functions as a transglutaminase enzyme, crosslinking fibrin (the primary protein in blood clots) to enhance its strength and stability. F13A1 is synthesized and secreted by platelets and monocytes/macrophages. It is activated by thrombin to execute its function in the final stages of clot formation. Beyond its role in blood clotting, F13A1 has been implicated in other biological processes, including wound healing and bone remodeling.
Factor XIII is activated by thrombin and calcium ions to a transglutaminase that catalyzes the formation of gamma-glutamyl-epsilon-lysine cross-links between fibrin chains, thereby stabilizing the fibrin clot. It also cross-links alpha-2-plasmin inhibitor, or fibronectin, to the alpha chains of fibrin.
F13A1 (Coagulation Factor XIII A1 Polypeptide) plays a crucial role in the final stage of the blood coagulation cascade. It functions as a transglutaminase enzyme that crosslinks fibrin (the main protein in blood clots) to create stronger, more stable clots. F13A1 is synthesized and secreted primarily by platelets and monocytes/macrophages and requires activation by thrombin to perform its crosslinking function. Beyond hemostasis, F13A1 participates in wound healing and bone remodeling processes .
The molecular structure of F13A1 is part of the larger Factor XIII complex, which exists in plasma as a heterotetramer composed of two A subunits with catalytic function and two B subunits that serve as carrier molecules. In platelets, Factor XIII consists solely of two A subunits (identical to those found in plasma) .
F13A1 monoclonal antibodies are typically developed through immunization and hybridoma technology. The process involves:
Immunizing mice with synthesized peptides derived from human F13A1
Extracting B cells from the spleens of immunized mice
Fusing these B cells with myeloma cells to create hybridomas
Screening and selecting hybridomas that produce F13A1-targeting antibodies
Culturing selected hybridomas in mouse abdominal cavities
Purifying the antibodies from ascites using affinity chromatography with specific immunogens
The resulting monoclonal antibodies demonstrate high specificity, with some validated across more than 19,000 full-length human proteins through techniques like HuProt™ Array . These antibodies typically recognize the 83kDa F13A1 protein and have been validated for various applications including ELISA and immunohistochemistry .
F13A1 monoclonal antibodies have been validated for multiple research applications:
| Application | Recommended Dilution | Notes |
|---|---|---|
| Immunohistochemistry (IHC) - Paraffin | 1:20-1:200 or 0.1-0.2 μg/ml | May require heat-induced epitope retrieval in Tris-EDTA buffer (pH 9.0) |
| ELISA | Varies by antibody | Validated for specific detection of human F13A1 |
| Protein Array | 1:100-1:2000 | For detection of F13A1 in complex protein mixtures |
These antibodies have demonstrated particular utility in dermatopathology, where they can differentiate between dermatofibroma (typically positive), dermatofibrosarcoma protuberans (variable/weak positive), and desmoplastic malignant melanoma (negative) .
When designing immunohistochemistry experiments with F13A1 monoclonal antibodies, the following controls are essential:
Positive tissue controls: Include tissues known to express F13A1, such as:
Negative tissue controls: Include tissues known to lack F13A1 expression or use tissues from F13A1 knockout models.
Antibody controls:
Primary antibody omission control
Isotype control (using an irrelevant antibody of the same isotype)
Concentration-matched non-specific antibody control
Antigen competition control: Pre-incubating the antibody with the immunizing peptide (aa46-181 of human F13A1 for some antibodies) should abolish specific staining .
Optimizing antibody concentration is critical, with recommended dilutions typically in the range of 1:20-1:200 or 0.1-0.2 μg/ml for paraffin sections, but this should be empirically determined for each experimental system .
A comprehensive validation strategy for F13A1 monoclonal antibodies in new experimental systems includes:
Specificity validation:
Western blot analysis to confirm binding to a protein of the expected molecular weight (83kDa)
Immunoprecipitation followed by mass spectrometry to confirm target identity
Testing in cell lines with known F13A1 expression levels
Testing in F13A1 knockout/knockdown systems
Application-specific validation:
For IHC: Optimize fixation, antigen retrieval (e.g., 45 min at 95°C in 10mM Tris with 1mM EDTA, pH 9.0), antibody concentration, and detection system
For ELISA: Determine optimal coating conditions, blocking reagents, antibody concentrations, and detection thresholds
For protein arrays: Validate signal-to-noise ratio and cross-reactivity
Batch-to-batch consistency checks:
Compare staining patterns between different lots
Maintain reference samples for comparison
Z-score and S-score assessment:
Proper validation ensures experimental reliability and reproducibility before embarking on full-scale studies.
Optimal sample preparation for F13A1 detection varies by tissue type and application:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin according to standard protocols
Cut sections at 4-5 μm thickness
For antigen retrieval, heat sections in 10mM Tris with 1mM EDTA, pH 9.0, for 45 min at 95°C, followed by cooling at room temperature for 20 minutes
Block endogenous peroxidase activity and non-specific binding sites
Apply F13A1 antibody at the optimized dilution (typically 0.1-0.2 μg/ml)
Snap-freeze tissues in liquid nitrogen or isopentane cooled in liquid nitrogen
Cut sections at 5-8 μm thickness
Fix in cold acetone or 4% paraformaldehyde
Apply F13A1 antibody at an optimized dilution
For adherent cells: Culture on chamber slides or coverslips
For suspension cells: Prepare cytospins
Fix with 4% paraformaldehyde or methanol
Permeabilize cells if detecting intracellular F13A1
Apply F13A1 antibody at the appropriate dilution
Different tissue types may require specific modifications to these protocols based on F13A1 expression levels and tissue characteristics.
F13A1 monoclonal antibodies provide powerful tools for investigating Factor XIII's role in various pathological conditions:
Use immunohistochemistry to assess F13A1 distribution and activation state in thrombi
Compare F13A1 levels and activity in normal versus pathological clots
Correlate F13A1 expression with thrombosis severity and clinical outcomes
Track F13A1-positive cells in normal versus impaired wound healing
Investigate the relationship between F13A1 expression and extracellular matrix organization
Study F13A1's interactions with other wound healing mediators
Examine F13A1 expression in inflammatory cell infiltrates
Investigate F13A1's role in tissue remodeling during chronic inflammation
Study potential F13A1-mediated crosslinking of inflammatory mediators
Utilize F13A1 antibodies to differentiate between histologically similar entities:
Investigate F13A1-positive dermal dendritic cells in various skin conditions
These applications contribute to understanding disease mechanisms and potentially identifying new therapeutic targets.
Combining F13A1 immunostaining with complementary techniques provides more comprehensive insights:
Co-stain for F13A1 alongside markers for:
Cell lineage (CD14, CD68 for monocytes/macrophages)
Activation states (CD80, CD86, CD163)
Other coagulation factors (thrombin, fibrin)
Analyze spatial relationships between F13A1-positive cells and other tissue components
Combine F13A1 immunostaining with transglutaminase activity assays
Use biotinylated substrate peptides to detect sites of active crosslinking
Correlate F13A1 protein presence with enzyme activity in tissue sections
Identify F13A1-positive cells or regions by immunostaining
Microdissect these areas for subsequent molecular analysis
Perform RNA-seq or proteomics on isolated material
Investigate protein-protein interactions involving F13A1
Detect in situ associations between F13A1 and potential substrates or regulatory proteins
Map the interactome of F13A1 in different tissue contexts
Apply F13A1 immunostaining to thick tissue sections or cleared tissues
Use confocal or light-sheet microscopy for volumetric analysis
Reconstruct the 3D distribution of F13A1-positive cells in complex tissues
These integrated approaches provide multidimensional data about F13A1's distribution, interactions, and functions in biological systems .
F13A1 monoclonal antibodies offer valuable approaches to investigate Factor XIII deficiency:
Use immunohistochemistry to quantify F13A1 protein levels in patient samples
Correlate protein expression with specific F13A1 gene mutations
Compare immunostaining patterns between type I deficiency (lacking both A and B subunits) and type II deficiency (lacking only A subunits)
Examine wound healing tissues from Factor XIII-deficient patients
Assess fibrin crosslinking patterns using F13A1 and fibrin co-staining
Investigate cellular compensation mechanisms in deficiency states
Track F13A1 levels before and after replacement therapy
Assess tissue distribution of exogenous Factor XIII in treated patients
Evaluate the relationship between F13A1 levels and clinical improvement
Study placental tissues in cases of habitual abortion associated with F13A1 deficiency
Examine F13A1 distribution in normal versus pathological placentas
Investigate potential mechanisms underlying pregnancy loss in deficiency states
This research helps bridge fundamental molecular understanding with clinical manifestations, potentially leading to improved diagnostic and therapeutic approaches for Factor XIII deficiency.
Researchers frequently encounter several challenges when working with F13A1 monoclonal antibodies:
Addressing these challenges requires systematic optimization and appropriate controls to ensure reliable and reproducible results .
Interpreting F13A1 expression patterns requires consideration of multiple factors:
Strong expression in:
Variable expression in:
Hepatocytes (typically lower levels)
Endothelial cells (context-dependent)
Assess whether staining represents zymogen (inactive) or activated F13A1
Activated F13A1 may show different subcellular distribution
Consider co-staining with activation markers
Compare expression patterns with established tissue references
Note changes in intensity, distribution, or cell type-specificity in pathological states
Consider quantitative assessment when comparing conditions
Develop consistent scoring criteria:
0: Negative (no staining)
1+: Weak (faint, barely perceptible staining)
2+: Moderate (distinct staining)
3+: Strong (intense staining)
Consider both percentage of positive cells and intensity
Use digital image analysis when possible for objective quantification
Accurate interpretation requires knowledge of F13A1 biology and function in different contexts, as well as careful consideration of technical factors that may influence staining patterns .
When analyzing F13A1 expression data across different experimental groups, several statistical approaches are appropriate:
For ordinal scoring systems (0, 1+, 2+, 3+):
Use non-parametric tests (Mann-Whitney U, Kruskal-Wallis) for between-group comparisons
Apply Spearman's rank correlation for association with other variables
For continuous measurements (% positive cells, staining intensity):
Apply parametric tests (t-test, ANOVA) if normality assumptions are met
Use appropriate transformations for non-normally distributed data
Apply multivariate statistical methods:
Principal Component Analysis (PCA) to identify patterns across multiple markers
Cluster analysis to identify subgroups with similar expression profiles
MANOVA for comparing multiple markers across groups
Kaplan-Meier survival analysis with log-rank tests for categorical F13A1 expression
Cox proportional hazards models for continuous measures or multivariable analysis
ROC curve analysis to determine optimal cutoff values for predictive purposes
Conduct power analysis to determine required sample sizes
For preliminary studies, consider the rule of thumb: at least 10-15 samples per group
For validation studies, larger sample sizes based on effect sizes from preliminary data
Appropriate statistical approach selection depends on study design, data distribution, and specific research questions. Consulting with a biostatistician during study design is highly recommended for complex study designs .
Development of next-generation F13A1 monoclonal antibodies could significantly advance Factor XIII research through:
Antibodies that selectively recognize active versus inactive F13A1
Tools that distinguish between free A subunits and those complexed with B subunits
Reagents that detect specific post-translational modifications
Higher-affinity antibodies for detecting low-level F13A1 expression
Antibodies optimized for challenging sample types (e.g., highly fibrous tissues)
Tools for detecting F13A1 in contexts previously difficult to study
Development of antibodies that recognize conserved epitopes across multiple species
Tools that enable translational research between animal models and human samples
Reagents validated for comparative studies across evolutionary lineages
Antibodies that modulate F13A1 activity for potential therapeutic applications
Tools for targeted delivery of payloads to F13A1-expressing cells
Reagents for monitoring F13A1 levels during therapeutic interventions
These advancements would provide researchers with more precise tools to dissect F13A1's roles in hemostasis, wound healing, bone remodeling, and pathological conditions, potentially leading to novel diagnostic and therapeutic approaches .
Several emerging technologies show promise for advancing F13A1 research:
Single-cell RNA-seq combined with F13A1 protein detection to correlate transcription and translation
Mass cytometry (CyTOF) incorporating F13A1 antibodies for high-dimensional phenotyping
Single-cell proteomics to study F13A1 in rare cell populations
Super-resolution microscopy for nanoscale visualization of F13A1 distribution
Intravital microscopy to track F13A1-positive cells in living organisms
Correlative light and electron microscopy to link F13A1 location with ultrastructural context
Spatial transcriptomics combined with F13A1 immunostaining
Imaging mass spectrometry for spatial mapping of F13A1 and its substrates
Multiplexed ion beam imaging (MIBI) for highly multiplexed protein detection
CRISPR screens to identify genes affecting F13A1 expression and function
Optogenetic tools to spatiotemporally control F13A1 activation
Engineered reporter systems to monitor F13A1 activity in real-time
Machine learning algorithms for automated analysis of F13A1 staining patterns
Integrative multi-omics approaches incorporating F13A1 protein data
Predictive modeling of F13A1 interactions and functions
These technologies will enable more comprehensive understanding of F13A1 biology across scales from molecular interactions to tissue-level functions .
F13A1 monoclonal antibodies have significant potential in translational applications:
Development of sensitive immunoassays for detecting F13A1 levels in blood
Creation of point-of-care tests for rapid assessment of Factor XIII deficiency
Incorporation into multiplex diagnostic panels for coagulation disorders
Tissue-based diagnostic markers for dermatopathology and other specialties
Identification of F13A1 expression patterns that correlate with disease outcomes
Development of standardized scoring systems with prognostic value
Integration with other biomarkers for improved risk stratification
Tools for monitoring Factor XIII replacement therapy efficacy
Assessment of tissue penetration and distribution of therapeutic Factor XIII
Evaluation of treatment response based on F13A1 levels and activity
Antibody-based modulation of F13A1 activity in thrombotic disorders
Targeted delivery of drugs to F13A1-expressing cells
F13A1-based imaging agents for visualizing thrombi or specific cell populations
Monitoring F13A1 in engineered tissues to assess functional maturation
Incorporation of F13A1 detection in quality control of tissue-engineered products
Assessment of crosslinking activity in biomaterial development
These translational applications could significantly impact clinical management of Factor XIII-related disorders and expand our therapeutic toolbox for conditions involving blood coagulation, wound healing, and tissue remodeling .