Property | Details | Source |
---|---|---|
Molecular Weight | 36 kDa | |
Biological Activity | 5,396 NIH Units/mg | |
Storage Conditions | -20°C to -80°C; avoid freeze-thaw cycles | |
Cleavage Specificity | Recognizes Leu-Val-Pro-Arg-Gly-Ser |
Recombinant thrombin is synthesized using HEK cells transfected with cDNA encoding prothrombin or modified prethrombin precursors . Key steps include:
Transfection: HEK 293, HEK 293T, or PER-C6 cells are engineered to express prothrombin variants.
Activation: Prothrombin is cleaved by proteases (e.g., factor Xa) during purification to yield active thrombin .
Glycosylation: HEK-derived thrombin exhibits human-like post-translational modifications, distinguishing it from CHO cell-produced variants .
Avoids animal-derived contaminants.
Achieves higher fidelity in protein folding and glycosylation compared to non-human cell lines .
Thrombin Generation Assays: Used to model intrinsic coagulation pathways in murine blood, demonstrating dose-dependent thrombin activity with contact activators like kaolin or polyphosphate .
Factor XII Activation: HEK-purified polyphosphate triggers thrombin generation via FXII-driven pathways, validated in F12−/− mice reconstituted with human FXII .
Fusion Protein Cleavage: The thrombin cleavage site (LVPRGS) is widely used to remove affinity tags from recombinant proteins .
Platelet Function Studies: Induces platelet activation and fibrin clot stabilization, mimicking physiological hemostasis .
Fibrin Sealants: Combined with recombinant fibrinogen for surgical hemostasis, offering reduced immunogenicity compared to bovine-derived products .
Cancer Research: Thrombin-mediated cleavage of platelet GARP releases TGF-β1, implicating it in tumor immune evasion mechanisms .
Mechanism: Directly converts fibrinogen to fibrin without intermediate factors, enabling rapid clot formation at wound sites .
Pharmacokinetics: Topical application limits systemic exposure; endogenous clearance involves hepatic degradation or thrombomodulin binding .
Human thrombin is a serine protease that plays a central role in the coagulation cascade, converting fibrinogen to fibrin and contributing to feedback amplification of coagulation. It also activates platelets, regulates coagulation through thrombomodulin-mediated effects, influences the fibrinolytic system via thrombin-activatable fibrinolysis inhibitor (TAFI), and directly affects clot structure . Beyond hemostasis, thrombin elicits responses in vascular endothelium, including shape and permeability changes and stimulation of cytokine production .
HEK293 expression systems provide significant advantages for human thrombin production because they ensure proper post-translational modifications and correct protein folding. The resulting recombinant enzyme has an identical amino acid sequence and similar structure to native human thrombin, greatly reducing immunogenicity concerns while maintaining full biological activity . Unlike bacterial expression systems, HEK cells properly perform human-specific post-translational modifications including glycosylation, γ-carboxylation, and correct disulfide bond formation.
Thrombin generation serves as a comprehensive measure of coagulation potential that accounts for both procoagulant and anticoagulant factors in the system. Measuring thrombin directly provides an evaluation of the total enzymatic work carried out by the coagulation cascade, making it a "global" coagulation assay . This approach offers several advantages over conventional coagulation tests:
It captures the amplification phase of coagulation, not just initiation
It reflects both procoagulant deficiencies and anticoagulant influences
It allows for the assessment of hypercoagulable states
It can potentially serve as a surrogate outcome for bleeding risk
Recombinant human thrombin (rhThrombin) offers several significant advantages over bovine thrombin (bThrombin) for research and clinical applications:
Characteristic | Recombinant Human Thrombin | Bovine Thrombin |
---|---|---|
Amino acid sequence | Identical to native human thrombin | Different from human thrombin |
Immunogenicity | Low - significantly fewer patients develop antibodies | Higher - more patients develop anti-thrombin antibodies |
Clinical efficacy | Achieves hemostasis within 10 minutes in 95.4% of surgical cases | Achieves hemostasis within 10 minutes in 95.1% of surgical cases |
Rapid action | >70% achieve hemostasis within 3 minutes | >70% achieve hemostasis within 3 minutes |
Safety concerns | Generally well-tolerated with minimal immunologic concerns | Potential for antibodies that cross-react with human factors |
In clinical trials, significantly fewer patients developed antibodies against rhThrombin compared to bThrombin after topical hemostatic treatment . In vascular surgery specifically, rhThrombin demonstrated hemostasis within 10 minutes at >90% of vascular anastomotic sites . Despite the higher production costs of rhThrombin, its equivalent efficacy and superior safety profile make it increasingly preferred for both research and clinical applications.
When designing experiments using human thrombin, researchers should adhere to several methodological principles:
Activity standardization: Determine and report thrombin activity using internationally recognized methods (chromogenic substrate assays with specific substrates like S-2238) rather than simply reporting protein concentration.
Storage and handling: Store concentrated thrombin solutions at -80°C in small aliquots to prevent repeated freeze-thaw cycles. Working solutions should be prepared fresh and kept on ice, as thrombin is prone to autodegradation at room temperature.
Buffer considerations: The activity of thrombin is highly dependent on pH, ionic strength, and calcium concentration. Use standardized buffers (typically HEPES or Tris-based, pH 7.4-7.5) for consistent results.
Contaminant awareness: Even high-purity preparations may contain trace amounts of other proteases or degradation products. Include appropriate controls (e.g., heat-inactivated thrombin, specific inhibitors like hirudin) to confirm thrombin-specific effects.
Concentration reporting: Report both the molar concentration and activity units (NIH units or IU) to facilitate comparison across studies.
Source documentation: Clearly document the source of thrombin (recombinant HEK-derived vs. plasma-derived), purity, and any modifications to the native sequence.
For quantitative experiments, developing dose-response curves rather than using single concentrations provides more robust data and accommodates variations in thrombin preparations between laboratories.
Optimizing TGAs with human thrombin as a reference standard requires attention to several critical factors:
Preanalytical considerations:
Sample collection method significantly impacts results, with citrate being the most common anticoagulant
Sample processing should ideally occur within 1-2 hours of collection to prevent ex vivo alterations in coagulation factors
Double centrifugation protocols (2,500g for 15 minutes, twice) are recommended to obtain platelet-poor plasma
Storage conditions impact stability, with -80°C recommended for long-term storage
Analytical considerations:
Calibration curves must be established using defined concentrations of the same human thrombin reference standard
Tissue factor concentration and source affect sensitivity (1-5 pM commonly used for plasma samples)
Phospholipid composition and concentration influence thrombin generation kinetics
Substrate selection impacts sensitivity and specificity
Continuous calibration should be employed to account for substrate consumption and inner filter effects
Data analysis approaches:
Multiple parameters should be reported beyond just Endogenous Thrombin Potential (ETP)
Statistical analysis should account for the interdependence of TGA parameters
Normalization to a reference plasma pool can reduce inter-assay variability
The interdependence of TGA parameters presents an analytical challenge, as selective reporting can lead to reporting bias and provide an incomplete picture of the data . When establishing a TGA protocol, researchers should carefully consider their specific research question to determine the most appropriate assay conditions and parameters to report.
Correlating in vitro thrombin generation with clinical bleeding outcomes faces several significant challenges:
Measurement complexities:
Clinical hemostasis is difficult to measure objectively and standardize across studies
The multifactorial nature of bleeding makes isolating thrombin's specific contribution complex
TGA parameters may be interdependent, making it difficult to identify which parameter best correlates with clinical outcomes
Selective reporting of specific TGA parameters (e.g., only ETP) can lead to reporting bias
Clinical correlation challenges:
Intraindividual bleed frequency is often low, requiring large sample sizes for meaningful correlations
Construct validity of current clinical hemostasis definitions has not been formally evaluated
The relative importance of kinetic versus quantitative TGA parameters remains uncertain
Bleeding may be influenced by factors beyond thrombin generation (vascular integrity, platelet function)
Methodological approaches to address these challenges:
Use of standardized bleeding assessment tools alongside TGA measurements
Longitudinal studies tracking both TGA parameters and bleeding events over time
Multivariate analysis controlling for known confounding factors
Subgroup analyses based on specific bleeding disorders or anticoagulation status
Although clinical criteria have been developed for adjudicating hemostasis in studies of hemostatic therapies, their construct validity has never been formally evaluated . This fundamental gap complicates efforts to establish thrombin generation as a reliable surrogate marker for bleeding risk.
The HEK expression system provides distinct advantages for human thrombin production compared to alternative expression platforms:
The HEK expression system's ability to perform these human-specific post-translational modifications results in recombinant human thrombin with structural and functional characteristics nearly identical to plasma-derived human thrombin. This is particularly important for:
Preserving the complex interaction network of thrombin with its substrates and regulatory proteins
Maintaining proper conformation of exosites I and II, which are critical for thrombin's specificity
Ensuring appropriate interaction with thrombomodulin and the protein C pathway
These advantages make HEK-derived thrombin particularly valuable for mechanistic studies requiring precise structural integrity and for translational research with direct clinical implications.
When designing experiments to study thrombin-mediated signaling in cellular models, researchers should consider:
Receptor considerations:
Expression levels of protease-activated receptors (PARs) vary significantly between cell types
Species differences exist in PAR activation and signaling, particularly between rodent and human systems
Receptor desensitization occurs following repeated thrombin exposure, requiring careful timing of stimulations
Potential cross-talk between thrombin receptors and other signaling pathways must be accounted for
Thrombin preparation:
Active site titration ensures consistent enzymatic activity between experiments
Glycosylation and other post-translational modifications impact receptor interactions
Storage conditions affect stability; activity should be verified before each experiment
Heat-inactivated controls should be included to distinguish between proteolytic and non-proteolytic effects
Experimental design:
Concentration-dependent effects should be evaluated (typical range: 0.1-10 nM for cellular signaling)
Time-course experiments capture both early and late signaling events
Serum-free conditions may be necessary to eliminate confounding effects of serum proteases
Specific thrombin inhibitors (e.g., hirudin, argatroban) should be included as controls
Analysis approaches:
Complementary methods should verify key findings (e.g., combining Western blotting with immunofluorescence)
Both canonical (PAR-mediated) and non-canonical thrombin signaling pathways should be considered
In vitro findings should be validated using physiologically relevant models when feasible
Pathway inhibitors can help delineate specific signaling cascades
A particularly valuable approach involves using a combined genetic and pharmacological strategy—comparing PAR knockout or knockdown cells with selective PAR antagonists to definitively identify receptor-specific contributions to observed signaling events.
Purification of recombinant human thrombin from HEK cell culture involves a multi-step process, with optimization at each stage enhancing final yield and purity:
Transfect HEK293 cells with a vector containing the human prothrombin gene (F2)
Use serum-free, chemically defined medium to simplify downstream purification
Include vitamin K (1-10 μg/mL) to ensure proper γ-carboxylation
Optimize culture conditions: 32-37°C, 5-8% CO2, harvesting 48-72 hours post-transfection
Clarify cell culture supernatant by centrifugation (3,000-5,000 × g, 15-30 minutes)
Filter through a 0.22 μm membrane to remove remaining debris
Apply to an affinity column appropriate for thrombin capture:
Benzamidine-Sepharose (for active thrombin)
Heparin-Sepharose (utilizes thrombin's exosite II)
Wash with high salt buffer (0.3-0.5 M NaCl) to remove weakly bound contaminants
If expressing prothrombin, controlled activation using purified factor Xa (0.5-1% w/w ratio to prothrombin)
Include calcium (5-10 mM) and phospholipids (20-50 μM)
Monitor activation by SDS-PAGE or enzymatic activity assays
Ion exchange chromatography:
Anion exchange (Q-Sepharose) at pH 7.4-8.0 or
Cation exchange (SP-Sepharose) at pH 5.5-6.5
Apply salt gradient elution (0-1 M NaCl)
Size exclusion chromatography to remove aggregates and degradation products
Hydrophobic interaction chromatography for removal of closely related impurities
Assess purity by SDS-PAGE (reducing and non-reducing conditions)
Determine specific activity using chromogenic substrates (S-2238)
Perform active site titration with known inhibitors
Typical yields from optimized processes range from 1-5 mg/L of culture medium with >95% purity by SDS-PAGE and specific activity of 2,000-3,000 NIH units/mg protein. Throughout purification, it's critical to include appropriate protease inhibitors and maintain optimal pH and temperature conditions to prevent autodegradation.
Standardization of thrombin activity measurements is crucial for comparing results across different experimental systems:
Reference standards:
Use internationally recognized standards (e.g., WHO International Standard for Thrombin)
Include internal laboratory standards with known activity relative to international standards
Regularly verify working standard activity against reference standards
Activity determination methods:
Chromogenic substrate assay:
Use S-2238 or other specific thrombin substrates
Standardize substrate concentration (typically 100-200 μM)
Control temperature (37°C) and pH (7.4-7.8)
Calculate activity based on initial velocity of substrate cleavage
Clotting-based assays:
Standardize fibrinogen concentration and source
Control temperature, pH, and ionic strength
Measure time to clot formation under defined conditions
Units of measurement:
Report in internationally recognized units:
NIH Units: historical standard based on clotting activity
IU (International Units): WHO standard
Enzymatic Units: nmol of substrate converted per minute
Include conversion factors between different unit systems
Active site titration:
Use irreversible inhibitors like PPACK or NPGB
Determine the stoichiometric endpoint of inhibition
Calculate the concentration of active enzyme
Use this as the definitive measure of active thrombin concentration
Standardization protocol:
Calibrate instruments and establish standard curves
Include multiple concentrations of thrombin in each assay
Verify linearity of response within the working range
Include positive and negative controls
Express results relative to the international standard
This standardized approach facilitates comparison across studies and enhances reproducibility in thrombin research, addressing a key challenge in the field.
Minimizing variability in thrombin generation assays is essential for reliable results in coagulation disorder studies:
Pre-analytical factors:
Standardize blood collection:
Use sodium citrate 3.2% as anticoagulant
Standardize needle gauge (19-21G recommended)
Establish consistent tourniquet time (<1 minute)
Discard first 3-5 mL of blood to avoid tissue factor contamination
Sample processing:
Process within 2 hours of collection
Use consistent centrifugation protocols (2,500g for 15 min twice for platelet-poor plasma)
Standardize plasma storage (-80°C) and thawing conditions (37°C water bath)
Analytical factors:
Reagent standardization:
Use same lot numbers where possible
Include calibrator with each run
Standardize tissue factor source and concentration
Use consistent phospholipid composition and concentration
Instrumentation:
Regular calibration of fluorometer or chromogenic detection systems
Temperature control (37°C ± 0.5°C)
Consistent plate reading intervals
Controls and normalization strategies:
Include reference plasma pool with each batch
Run samples in duplicate or triplicate
Consider normalization to reference plasma results
Use internal quality control samples with known TGA parameters
Specific strategies for coagulation disorders:
Hemophilia studies:
Standardize factor replacement levels before testing
Include factor-specific depleted plasma controls
Anticoagulation studies:
Standardize timing of sample collection relative to drug administration
Include drug calibrators at known concentrations
By implementing these strategies, researchers can substantially reduce coefficients of variation from >20% to <10% for most TGA parameters , enhancing the reliability of results particularly in studies of coagulation disorders.
Effectively comparing results from different thrombin generation assay platforms requires careful consideration of methodological differences and standardization approaches:
Platform comparison table:
Platform Characteristic | Calibrated Automated Thrombogram | ST Genesia | Technothrombin TGA | Custom Research Methods |
---|---|---|---|---|
Detection method | Fluorogenic | Fluorogenic | Fluorogenic/Chromogenic | Various |
Calibration approach | Thrombin calibrator | Thrombin calibrator | Reference curve | Method-dependent |
Typical substrate | Z-Gly-Gly-Arg-AMC | Z-Gly-Gly-Arg-AMC | Various | Method-dependent |
Continuous calibration | Yes | Yes | Method-dependent | Method-dependent |
α2M correction | Yes | Yes | Method-dependent | Method-dependent |
Parameter calculation | Specialized software | Automated | Specialized software | Method-dependent |
Standardization approaches:
Use of reference plasmas:
Analyze identical reference plasmas across platforms
Develop conversion factors based on reference plasma results
Express results as ratios to reference plasma values
External quality control programs:
Participate in external quality assessment schemes
Use results to harmonize between platforms
Standard additions method:
Spike samples with known quantities of calibrated thrombin
Compare recovery and response across platforms
Reporting recommendations:
Provide detailed methodological descriptions
Report all assay conditions (tissue factor, phospholipids, etc.)
Include raw data when possible alongside calculated parameters
Specify the calculation methods used for deriving parameters
Report both absolute values and normalized results
Cross-validation approach:
Select a subset of samples representing the full range of expected results
Analyze these samples on all platforms being compared
Establish correlation coefficients and conversion equations
Validate these equations with a new set of samples
Report both the original and converted values when publishing
While absolute values will differ between platforms, relative changes and trends can be comparable if standardization approaches are implemented . Researchers should emphasize consistent use of a single platform within a study whenever possible to minimize variability.
Recombinant human thrombin from HEK cells offers several distinctive advantages compared to plasma-derived human thrombin in research applications:
Characteristic | Recombinant Human Thrombin (HEK) | Plasma-Derived Human Thrombin |
---|---|---|
Source | HEK293 cell expression system | Human plasma fractionation |
Purity | Typically >95% | Variable (80-95%) |
Batch-to-batch consistency | High | Moderate to low |
Risk of contamination | Low (no human-derived materials) | Potential risk of bloodborne pathogens |
Post-translational modifications | Complete human-type modifications | Native human modifications |
Specific activity | 2,000-3,000 NIH units/mg | 2,000-3,000 NIH units/mg |
α-thrombin vs. degradation forms | Predominantly α-thrombin | Contains α-thrombin and various degradation products |
Research applications advantage | Defined composition ideal for mechanistic studies | Better represents the heterogeneity found in vivo |
Key considerations for research applications:
Mechanistic studies benefit from the defined composition of recombinant thrombin
Clinical correlation studies may benefit from plasma-derived thrombin's heterogeneity
Availability and consistency favor recombinant products for long-term research programs
The recombinant human thrombin shares an identical amino acid sequence with native human protein and demonstrates similar efficacy in clinical applications . For most research applications, the advantages of recombinant human thrombin (consistency, safety, defined composition) outweigh the potential benefits of plasma-derived thrombin, particularly for mechanistic studies requiring precise control of experimental variables.
When investigating thrombin's signaling functions beyond coagulation, researchers should consider:
Receptor biology considerations:
Cell-specific PAR expression profiles vary significantly between tissue and cell types
PAR-1, PAR-3, and PAR-4 serve as thrombin receptors with different activation thresholds
PAR receptor desensitization and internalization kinetics affect repeated exposures
Non-PAR mediated effects may occur through alternate mechanisms
Experimental design considerations:
Concentration range selection:
Low concentrations (50-100 pM) may selectively activate PAR-1
Higher concentrations (>1 nM) activate multiple PARs
Very high concentrations (>10 nM) may have non-specific effects
Temporal dynamics:
Immediate signaling (seconds to minutes): calcium flux, phosphorylation events
Intermediate responses (minutes to hours): gene expression changes
Long-term effects (hours to days): phenotypic alterations, cell fate decisions
Controls to include:
PAR-specific activating peptides to distinguish receptor-specific effects
Catalytically inactive thrombin (e.g., PPACK-thrombin) to identify non-proteolytic effects
Receptor antagonists or genetic knockdowns to confirm receptor dependency
Advanced approaches:
Microfluidic systems to model physiological flow conditions
3D tissue models that better recapitulate in vivo cellular architecture
Live-cell imaging with fluorescent biosensors to track signaling dynamics
Single-cell analysis to capture heterogeneity in cellular responses
Common research applications:
Endothelial barrier function and vascular permeability
Inflammatory responses in various cell types
Neural development and synaptic plasticity
Tumor cell migration and metastasis
Fibroblast activation and tissue remodeling
When publishing results, researchers should clearly specify the thrombin concentration, exposure duration, cell type, culture conditions, and thrombin source to facilitate reproducibility and cross-study comparisons.
Thrombin generation assays (TGAs) provide valuable insights when studying novel anticoagulants and hemostatic agents:
Applications for anticoagulant development:
Mechanism characterization:
Direct thrombin inhibitors primarily affect peak height and ETP
Factor Xa inhibitors primarily affect lag time and time to peak
Vitamin K antagonists affect all parameters
Dose-response relationship determination:
Establish EC50 values for thrombin generation inhibition
Identify therapeutic windows that balance anticoagulant effect with bleeding risk
Compare potency across different anticoagulant classes
Reversal agent evaluation:
Quantify restoration of thrombin generation parameters
Determine dose requirements for effective reversal
Assess duration of reversal effect
Applications for hemostatic agent development:
Experimental design considerations:
Trigger selection:
Low tissue factor (1-5 pM) for sensitive detection of anticoagulant effects
Multiple trigger concentrations to assess pathway-specific effects
Addition of thrombomodulin to evaluate protein C pathway interactions
Sample matrices:
Normal pooled plasma for standard testing
Factor-deficient plasmas for targeted investigations
Patient plasmas to assess individual variability
Control inclusion:
Standard curve of unfractionated heparin as a reference anticoagulant
Normal reference plasma with each batch
Pre- and post-treatment samples for paired analysis
TGAs can identify subtle differences between anticoagulants or hemostatic agents that might not be detected by conventional coagulation tests, making them particularly valuable for comparative studies and mechanism elucidation .
Several cutting-edge technologies are transforming thrombin research:
Advanced imaging technologies:
Intravital microscopy allows real-time visualization of thrombin activity in living animals
Fluorescence resonance energy transfer (FRET)-based thrombin sensors enable spatial and temporal mapping
Super-resolution microscopy reveals nanoscale distribution of thrombin within fibrin networks
4D imaging captures dynamic changes in thrombin activity during clot formation and resolution
Biosensor technologies:
Designed ankyrin repeat proteins (DARPins) can be engineered as highly specific thrombin sensors
Customized advanced modular bispecific extracellular receptors (AMBERs) combine with generalized extracellular molecule sensors (GEMS) for detecting thrombin-related products
Microfluidic devices model vascular environments and measure thrombin generation under flow
Aptamer-based sensors provide highly specific thrombin detection in complex biological matrices
Systems biology approaches:
Computational models integrate thrombin's multiple roles in coagulation networks
Multi-omics studies correlate thrombin activity with proteomic and transcriptomic changes
Machine learning algorithms predict thrombin generation from clinical parameters
Network analysis of thrombin-mediated signaling pathways in various cell types
Genetic engineering applications:
CRISPR/Cas9 modification of thrombin-related genes to study specific domains
Knock-in models with fluorescently tagged thrombin for in vivo tracking
Designer cell lines that detect pathological concentrations of fibrin degradation products (related to thrombin activity) and respond with production of therapeutic anti-thrombotic proteins
These technologies are revealing new insights into:
The spatial and temporal dynamics of thrombin activity during clot formation
Cell type-specific responses to thrombin signaling
The integration of thrombin into broader hemostatic and inflammatory networks
Structure-function relationships in thrombin's interactions with various substrates and receptors
Thrombin research is driving advances in personalized medicine approaches for coagulation disorders:
Precision diagnostics:
Individualized thrombin generation profiles serve as "coagulation fingerprints"
Integration of thrombin generation data with genetic information identifies personalized risk factors
Machine learning algorithms predict individual bleeding or thrombotic risk based on thrombin dynamics
Point-of-care thrombin generation testing enables rapid personalization of therapy
Therapeutic tailoring approaches:
Dose adjustment of anticoagulants based on individual thrombin generation response
Selection of optimal hemostatic agent based on ex vivo thrombin generation testing
Prediction of reversal agent efficacy for specific patients
Monitoring of treatment response through serial thrombin generation measurements
Emerging personalized applications:
Hemophilia treatment:
Using thrombin generation to optimize factor replacement dosing
Identifying patients who may benefit from non-factor therapies
Predicting inhibitor development risk
Anticoagulation management:
Identifying optimal drug and dose for individual patients
Predicting bleeding risk during invasive procedures
Guiding reversal strategies during bleeding events
Perioperative management:
Customizing hemostatic agent selection based on individual deficits
Predicting transfusion requirements
Monitoring for hypercoagulable states post-procedure
Thrombin is a serine protease enzyme that plays a crucial role in the blood coagulation process. It is derived from prothrombin, a glycoprotein produced in the liver. Thrombin’s primary function is to convert fibrinogen into fibrin, leading to blood clot formation. The recombinant form of thrombin, expressed in Human Embryonic Kidney (HEK) 293 cells, is widely used in research and therapeutic applications due to its high purity and activity.
HEK 293 cells are immortalized human embryonic kidney cells that are commonly used for protein production. These cells are robust, fast-growing, and easy to maintain, making them ideal for producing recombinant proteins. The recombinant human thrombin produced in HEK 293 cells is a glycoprotein heterodimer, consisting of a heavy chain (31 kDa) and a light chain (6 kDa) joined by a disulfide bond .
Thrombin is composed of two chains: the A chain (light chain) and the B chain (heavy chain). The enzyme selectively cleaves the Arg-Gly bonds in fibrinogen to form fibrin and release fibrinopeptides A and B . The predominant form of thrombin in vivo is the zymogen prothrombin (factor II), which is activated by factor Xa in the presence of factor Va, calcium ions, and phospholipids .
Recombinant thrombin is used in various applications, including:
The use of recombinant thrombin expressed in HEK 293 cells offers several advantages: